Pancreatic ductal adenocarcinoma (PDAC)

胰腺导管腺癌 (PDAC)
  • 文章类型: Journal Article
    仅根据个别并发症评估胰十二指肠切除术(PD)的围手术期结果并不全面,围手术期结局与胰腺导管腺癌(PDAC)患者长期预后之间的关系尚不确定.我们的研究旨在评估一种新型复合指标的影响,教科书成果(TO),PDAC治疗PD患者的长期预后。
    本研究对2018年1月至2021年12月在我院因病理证实的PDAC而接受PD的139例患者进行了回顾性分析。应用排除标准后,共有111例患者被纳入后续分析.这些患者分为两组:非TO组(n=42)和TO组(n=69)。采用Kaplan-Meier存活曲线来描述TO与无病生存期(DFS)和总生存期(OS)之间的关系。采用Cox回归评估达到TO对长期生存的影响。采用Logistic回归分析影响TO达成的危险因素。
    在111名PDAC患者中,PD后达到69(62.2%)。TO的实现显着改善了PDAC患者的OS[P=0.03;风险比(HR)=0.60;95%置信区间(CI):0.37-0.83]。Cox回归分析表明,达到TO是OS的保护因素(P=0.04;HR=4.08;95%CI:1.07-15.61)。Logistic回归分析显示,术后第3天引流液中高淀粉酶(>1,300U/L)不利于达到TO[比值比(OR)=0.10;95%CI:0.02-0.58;P=0.01],较长的手术持续时间(≥6.25小时)不利于达到TO(OR=0.19;95%CI:0.06-0.54;P=0.002),和软胰腺质地不利于实现TO(OR=0.31;95%CI:0.10-0.93,P=0.04)。
    实现TO可显着改善PDAC患者的OS,并有可能作为稳健的预后指标。展望未来,成为医院标准的手术质量控制措施是非常必要的。
    UNASSIGNED: Assessing the perioperative outcomes of pancreaticoduodenectomy (PD) based solely on individual complications is not comprehensive, and the association between perioperative outcomes and the long-term prognosis of individuals diagnosed with pancreatic ductal adenocarcinoma (PDAC) remains uncertain. Our study is designed to evaluate the impact of a novel composite indicator, textbook outcomes (TO), on the long-term prognosis of patients undergoing PD for PDAC.
    UNASSIGNED: This study conducted a retrospective analysis of 139 patients who underwent PD for pathologically confirmed PDAC at our hospital between January 2018 and December 2021. After applying exclusion criteria, a total of 111 patients were included in the subsequent analysis. These patients were categorized into two groups: the non-TO group (n=42) and the TO group (n=69). The Kaplan-Meier survival curve was employed to describe the relationship between TO and disease-free survival (DFS) and overall survival (OS). Cox regression was employed to assess the impact of achieving TO on long-term survival. Logistic regression was employed to investigate the risk factors affecting the achievement of TO.
    UNASSIGNED: Out of the 111 PDAC patients, 69 (62.2%) achieved TO following PD. The achievement of TO significantly improved the OS of PDAC patients [P=0.03; hazard ratio (HR) =0.60; 95% confidence interval (CI): 0.37-0.83]. Cox regression analysis indicated that achieving TO was a protective factor for OS (P=0.04; HR =4.08; 95% CI: 1.07-15.61). Logistic regression analysis indicated that high amylase in drainage fluid on the third day after surgery (>1,300 U/L) was detrimental to achieve TO [odds ratio (OR) =0.10; 95% CI: 0.02-0.58; P=0.01], longer surgery durations (≥6.25 hours) was detrimental to achieve TO (OR =0.19; 95% CI: 0.06-0.54; P=0.002), and soft pancreatic texture was detrimental to achieve TO (OR =0.31; 95% CI: 0.10-0.93, P=0.04).
    UNASSIGNED: Achievement of TO significantly improves the OS of PDAC patients and has the potential to serve as a robust prognostic indicator. Looking ahead, it is highly necessary for TO to become a standard surgical quality control measure in hospitals.
    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
    胰腺导管腺癌(PDAC)患者的区域淋巴结(LN)清扫没有统一的范围。不完整的局部LN夹层可导致术后复发,而区域LN夹层范围的盲目扩大会显着增加围手术期风险,而不会显着延长总生存期。我们旨在建立一种基于双层探测器能谱计算机断层扫描(DLCT)的无创可视化工具,以预测PDAC患者发生局部LN转移的可能性。
    对总共163个区域性LN进行了审查,并将其分为转移性队列(n=58个LN)和非转移性队列(n=105个LN)。在两个队列之间比较了DLCT定量参数以及区域LN的最长轴与最短轴(L/S)的节点比。DLCT定量参数包括动脉期碘浓度(APIC),动脉期标准化碘浓度(APNIC),动脉期有效原子序数(APZeff),动脉期归一化有效原子序数(APNZeff),动脉期频谱衰减曲线的斜率(APλHU),门静脉期碘浓度(PVPIC),门静脉期标准化碘浓度(PVPNIC),门静脉阶段的有效原子序数(PVPZeff),门静脉期归一化有效原子序数(PVPNZeff),以及门静脉期(PVPλHU)的光谱衰减曲线的斜率。采用基于曲线下面积(AUC)的Logistic回归分析对有意义的DLCT定量参数的诊断性能,L/S,以及将重要的DLCT定量参数和L/S相结合的模型。开发了基于具有最高诊断性能的模型的列线图作为预测指标。通过校准曲线和决策曲线分析(DCA)评估列线图的拟合优度和临床适用性。
    APNIC+L/S(APNIC+L/S)的组合型号在所有型号中具有最高的诊断性能,产生AUC,灵敏度,和0.878的特异性[95%置信区间(CI):0.825-0.931],分别为0.707和0.886。校准曲线表明APNIC-L/S列线图在预测概率和实际概率之间具有良好的一致性。同时,决策曲线表明,APNIC-L/S列线图可以产生比全部或不干预策略更大的净收益,阈值概率范围从0.0到0.75。
    作为一种有效且可视的非侵入性预测工具,在PDAC患者中,APNIC-L/S列线图对鉴别转移性LN具有良好的预测功效.
    UNASSIGNED: There is no unified scope for regional lymph node (LN) dissection in patients with pancreatic ductal adenocarcinoma (PDAC). Incomplete regional LN dissection can lead to postoperative recurrence, while blind expansion of the scope of regional LN dissection significantly increases the perioperative risk without significantly prolonging overall survival. We aimed to establish a noninvasive visualization tool based on dual-layer detector spectral computed tomography (DLCT) to predict the probability of regional LN metastasis in patients with PDAC.
    UNASSIGNED: A total of 163 regional LNs were reviewed and divided into a metastatic cohort (n=58 LNs) and nonmetastatic cohort (n=105 LNs). The DLCT quantitative parameters and the nodal ratio of the longest axis to the shortest axis (L/S) of the regional LNs were compared between the two cohorts. The DLCT quantitative parameters included the iodine concentration in the arterial phase (APIC), normalized iodine concentration in the arterial phase (APNIC), effective atomic number in the arterial phase (APZeff), normalized effective atomic number in the arterial phase (APNZeff), slope of the spectral attenuation curves in the arterial phase (APλHU), iodine concentration in the portal venous phase (PVPIC), normalized iodine concentration in the portal venous phase (PVPNIC), effective atomic number in the portal venous phase (PVPZeff), normalized effective atomic number in the portal venous phase (PVPNZeff), and slope of the spectral attenuation curves in the portal venous phase (PVPλHU). Logistic regression analysis based on area under the curve (AUC) was used to analyze the diagnostic performance of significant DLCT quantitative parameters, L/S, and the models combining significant DLCT quantitative parameters and L/S. A nomogram based on the models with highest diagnostic performance was developed as a predictor. The goodness of fit and clinical applicability of the nomogram were assessed through calibration curve and decision curve analysis (DCA).
    UNASSIGNED: The combined model of APNIC + L/S (APNIC + L/S) had the highest diagnostic performance among all models, yielding an AUC, sensitivity, and specificity of 0.878 [95% confidence interval (CI): 0.825-0.931], 0.707, and 0.886, respectively. The calibration curve indicated that the APNIC-L/S nomogram had good agreement between the predicted probability and the actual probability. Meanwhile, the decision curve indicated that the APNIC-L/S nomogram could produce a greater net benefit than could the all- or-no-intervention strategy, with threshold probabilities ranging from 0.0 to 0.75.
    UNASSIGNED: As a valid and visual noninvasive prediction tool, the APNIC-L/S nomogram demonstrated favorable predictive efficacy for identifying metastatic LNs in patients with PDAC.
    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
    由于缺乏早期诊断方法和有效药物,胰腺导管腺癌(PDAC)预后极差.DNA甲基化,转录组表达和基因拷贝数变异(CNV)与各种疾病的发生和发展有着重要的关系。这项研究的目的是筛选可靠的早期诊断生物标志物和潜在的药物的基础上整合的多组分析。
    我们使用甲基化,转录组和CNV谱建立PDAC诊断模型。使用PDAC样品外部验证三个模型相关基因的蛋白质表达。然后,PDAC的潜在治疗药物通过与现有药物和基因相关的相互作用信息进行鉴定.
    从589个常见DMRs中选择了四个显着的差异甲基化区域(DMRs),以建立PDAC的高性能诊断模型。然后,四个枢纽基因,获得PHF12、FXYD3、PRKCB和ZNF582。外部验证结果显示,与癌旁正常组织相比,肿瘤组织中PHF12、FXYD3和PRKCB蛋白表达水平均上调(P<0.05)。通过在线数据集的基因表达分析,筛选并重新利用具有抗PDAC活性的有希望的候选药物。五种药物,包括托普替康,PD-0325901,帕比司他,紫杉醇和17-AAG,在27个PDAC细胞系中活性最高的被过滤。
    总的来说,基于4个显著DMRs建立的诊断模型能准确区分肿瘤组织和正常组织。五种候选药物可能被重新用作特定PDAC患者的有希望的治疗剂。
    UNASSIGNED: Due to a lack of early diagnosis methods and effective drugs, pancreatic ductal adenocarcinoma (PDAC) has an extremely poor prognosis. DNA methylation, transcriptome expression and gene copy number variation (CNV) have critical relationships with development and progression of various diseases. The purpose of the study was to screen reliable early diagnostic biomarkers and potential drugs based on integrative multiomics analysis.
    UNASSIGNED: We used methylation, transcriptome and CNV profiles to build a diagnostic model for PDAC. The protein expression of three model-related genes were externally validated using PDAC samples. Then, potential therapeutic drugs for PDAC were identified by interaction information related to existing drugs and genes.
    UNASSIGNED: Four significant differentially methylated regions (DMRs) were selected from 589 common DMRs to build a high-performance diagnostic model for PDAC. Then, four hub genes, PHF12, FXYD3, PRKCB and ZNF582, were obtained. The external validation results showed that PHF12, FXYD3 and PRKCB protein expression levels were all upregulated in tumor tissues compared with adjacent normal tissues (P<0.05). Promising candidate drugs with activity against PDAC were screened and repurposed through gene expression analysis of online datasets. The five drugs, including topotecan, PD-0325901, panobinostat, paclitaxel and 17-AAG, with the highest activity among 27 PDAC cell lines were filtered.
    UNASSIGNED: Overall, the diagnostic model built based on four significant DMRs could accurately distinguish tumor and normal tissues. The five drug candidates might be repurposed as promising therapeutics for particular PDAC 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
    胰腺导管腺癌(PDAC),占胰腺癌(PC)的绝大多数,是一种高度侵袭性的恶性肿瘤,预后不佳。年龄被证明是影响PDAC患者生存结果的独立因素。我们的研究旨在确定预后因素并构建列线图来预测年龄≥60岁的PDAC患者的生存。
    从监测中收集年龄≥60岁的PDAC患者的数据,流行病学,和结束结果(SEER)数据库。多因素Cox回归分析用于确定总生存期(OS)和癌症特异性生存期(CSS)的预后因素。并通过校准图构建和验证了两个列线图,一致性指数(C指数)和决策曲线分析(DCA)。此外,温州医科大学附属第一医院432例患者作为外部队列。应用Kaplan-Meier曲线进一步验证列线图的临床有效性。
    确定了十个独立的预后因素来建立列线图。基于OS列线图的训练组和验证组的C指数分别为0.759和0.760,高于肿瘤淋巴结转移(TNM)分期系统的C指数(分别为0.638和0.636)。校准曲线显示预测和观察之间的高度一致性。与TNM系统相比,还获得了更好的接收器操作特征(ROC)曲线下面积(AUC)值和DCA。基于列线图的风险分层可以区分具有不同生存风险的患者。
    我们构建并外部验证了年龄≥60岁的PDAC患者的基于人群的生存预测列线图。新模型可以帮助临床医生个性化生存预测和风险评估。
    UNASSIGNED: Pancreatic ductal adenocarcinoma (PDAC), which accounts for the vast majority of pancreatic cancer (PC), is a highly aggressive malignancy with a dismal prognosis. Age is shown to be an independent factor affecting survival outcomes in patients with PDAC. Our study aimed to identify prognostic factors and construct a nomogram to predict survival in PDAC patients aged ≥60 years.
    UNASSIGNED: Data of PDAC patients aged ≥60 years were collected from the Surveillance, Epidemiology, and End Results (SEER) database. Multivariate Cox regression analysis was used to determined prognostic factors of overall survival (OS) and cancer-specific survival (CSS), and two nomograms were constructed and validated by calibration plots, concordance index (C-index) and decision curve analysis (DCA). Additionally, 432 patients from the First Affiliated Hospital of Wenzhou Medical University were included as an external cohort. Kaplan-Meier curves were applied to further verify the clinical validity of the nomograms.
    UNASSIGNED: Ten independent prognostic factors were identified to establish the nomograms. The C-indexes of the training and validation groups based on the OS nomogram were 0.759 and 0.760, higher than those of the tumor-node-metastasis (TNM) staging system (0.638 and 0.636, respectively). Calibration curves showed high consistency between predictions and observations. Better area under the receiver operator characteristic (ROC) curve (AUC) values and DCA were also obtained compared to the TNM system. The risk stratification based on the nomogram could distinguish patients with different survival risks.
    UNASSIGNED: We constructed and externally validated a population-based survival-predicting nomogram for PDAC patients aged ≥60 years. The new model could help clinicians personalize survival prediction and risk assessment.
    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:胰腺导管腺癌(PDAC)以其强大而致命的性质而闻名,在恶性肿瘤中声名狼藉.由于其具有挑战性的早期诊断,恶性程度高,以及对化疗药物的耐药性,在肿瘤学领域,胰腺癌的治疗一直非常困难。γ-谷氨酰环基转移酶(GGCT),谷胱甘肽代谢中的一种重要酶,与几种肿瘤类型的增殖和进展有关,而GGCT在胰腺导管腺癌中的生物学功能尚不清楚。
    方法:通过蛋白质印迹验证GGCT的表达谱,免疫组织化学,和RT-qPCR在胰腺癌组织样品和细胞系。功能富集分析,包括GSVA,ssGSEA,GO,和KEGG进行了研究,以探讨GGCT的生物学作用。此外,CCK8,Edu,菌落形成,迁移,并采用侵袭试验评价GGCT对胰腺癌细胞增殖和迁移能力的影响。此外,利用LASSO机器学习算法建立与GGCT相关的预后模型.
    结果:我们的研究表明,GGCT在胰腺癌组织和细胞中的表达增加,提示与患者预后较差有关。此外,我们探索了GGCT在泛癌症和胰腺癌环境中的免疫调节作用,发现GGCT在各种类型的肿瘤中可能与免疫抑制调节有关。具体来说,在胰腺癌中GGCT高表达的患者中,各种免疫细胞的浸润减少,导致对免疫疗法的反应性较差,生存率较差。体内和体外实验表明,GGCT的下调可显着抑制胰腺癌细胞的增殖和转移。此外,这种抑制作用似乎与GGCT对c-Myc的调节有关。基于来自GGCT的基因构建了一个预后模型,证明了良好的生存预后和免疫疗法反应的强大预测能力。
    BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is renowned for its formidable and lethal nature, earning it a notorious reputation among malignant tumors. Due to its challenging early diagnosis, high malignancy, and resistance to chemotherapy drugs, the treatment of pancreatic cancer has long been exceedingly difficult in the realm of oncology. γ-Glutamyl cyclotransferase (GGCT), a vital enzyme in glutathione metabolism, has been implicated in the proliferation and progression of several tumor types, while the biological function of GGCT in pancreatic ductal adenocarcinoma remains unknown.
    METHODS: The expression profile of GGCT was validated through western blotting, immunohistochemistry, and RT-qPCR in both pancreatic cancer tissue samples and cell lines. Functional enrichment analyses including GSVA, ssGSEA, GO, and KEGG were conducted to explore the biological role of GGCT. Additionally, CCK8, Edu, colony formation, migration, and invasion assays were employed to evaluate the impact of GGCT on the proliferation and migration abilities of pancreatic cancer cells. Furthermore, the LASSO machine learning algorithm was utilized to develop a prognostic model associated with GGCT.
    RESULTS: Our study revealed heightened expression of GGCT in pancreatic cancer tissues and cells, suggesting an association with poorer patient prognosis. Additionally, we explored the immunomodulatory effects of GGCT in both pan-cancer and pancreatic cancer contexts, found that GGCT may be associated with immunosuppressive regulation in various types of tumors. Specifically, in patients with high expression of GGCT in pancreatic cancer, there is a reduction in the infiltration of various immune cells, leading to poorer responsiveness to immunotherapy and worse survival rates. In vivo and in vitro assays indicate that downregulation of GGCT markedly suppresses the proliferation and metastasis of pancreatic cancer cells. Moreover, this inhibitory effect appears to be linked to the regulation of GGCT on c-Myc. A prognostic model was constructed based on genes derived from GGCT, demonstrating robust predictive ability for favorable survival prognosis and response to 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
    溶质载体家族16成员1(SLC16A1)在许多类型的癌症中充当生物标志物。肿瘤免疫浸润在肿瘤进展和治疗中越来越受到重视。我们的研究目的是探讨SLC16A1与胰腺导管腺癌(PDAC)中肿瘤免疫微环境之间的关系。
    数据来自癌症基因组图谱。使用xCell网工具根据SLC16A1表达计算免疫细胞的比例。为进一步探讨SLC16A1的作用机制,通过加权基因共表达网络分析,从差异表达基因中筛选免疫相关基因。通过基因本体论和京都百科全书的基因和基因组分析,筛选采用单变量Cox回归和最小绝对收缩率与选择算子回归模型相结合的相关性分析(P<0.05)。接下来,应用BERSORTCI剖析免疫细胞与5个重要基因的相干性。通过免疫组织化学染色实验阐明了SLC16A1在胰腺癌中的表达及其临床作用。最后,通过体外实验评价SLC16A1对肿瘤免疫结果的影响。
    SLC16A1在PDAC组织中过度表达,可能是一个独立的预后因素。SLC16A1与总生存期呈显著负相关,并抑制PDAC的肿瘤免疫。在临床上,SLC16A1表达与肿瘤进展和不良预后呈显著正相关。我们还发现SLC16A1可以抑制CD8T细胞的抗肿瘤能力。
    SLC16A1是PDAC预后的生物标志物,可以影响PDAC的免疫环境。这些发现为PDAC的治疗提供了新的见解。
    UNASSIGNED: Solute carrier family 16 member 1 (SLC16A1) serves as a biomarker in numerous types of cancer. Tumor immune infiltration has drawn increasing attention in cancer progression and treatment. The objective of our study was to explore the association between SLC16A1 and the tumor immune microenvironment in pancreatic ductal adenocarcinoma (PDAC).
    UNASSIGNED: Data were obtained from The Cancer Genome Atlas. The xCell web tool was used to calculate the proportion of immune cells according to SLC16A1 expression. To further explore the mechanism of SLC16A1, immunity-related genes were screened from differentially expressed genes through weighted gene coexpression network analysis, examined via Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses, and filtrated using univariate Cox regression and least absolute shrinkage and selection operator regression model combined correlation analysis (P<0.05). Next, CIBERSORT was used to analyze the correlation between immune cells and five important genes. SLC16A1 expression and its clinical role in pancreatic cancer was clarified via immunohistochemical staining experiments. Finally, the effects of SLC16A1 on the results of cancer immunity were evaluated by in vitro experiments.
    UNASSIGNED: SLC16A1 was overexpressed in PDAC tissues and could be an independent prognostic factor. SLC16A1 was significantly negatively correlated with overall survival and suppressed the tumor immunity of PDAC. In clinic, SLC16A1 expression was significantly positively correlated with tumor progression and poor prognosis. We also found that SLC16A1 could suppress the antitumor ability of CD8+ T cells.
    UNASSIGNED: SLC16A1 is a biomarker for the prognosis of PDAC and can influence the immune environment of PDAC. These findings provide new insights into the treatment of PDAC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 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

       PDF(Pubmed)

  • 文章类型: Journal Article
    胰腺癌,尤其是胰腺导管腺癌(PDAC),是美国癌症相关死亡的第四大原因,具有挑战性的治疗和令人沮丧的预后。随着免疫疗法成为减轻PDAC恶性进展的有希望的途径,全面了解肿瘤的免疫抑制特性成为当务之急。本文系统地研究了PDAC内部复杂的免疫抑制网络,强调了低氧酸性胰腺肿瘤微环境中免疫抑制细胞与因子之间的显著串扰。通过阐明这些机制,我们的目标是提供潜在的免疫治疗策略和治疗目标的见解,为未来PDAC免疫抑制研究奠定基础。认识到免疫抑制对PDAC侵袭和转移的深远影响,本讨论旨在促进PDAC患者更有效和有针对性的免疫疗法的发展.
    Pancreatic cancer, particularly pancreatic ductal adenocarcinoma (PDAC), stands as the fourth leading cause of cancer-related deaths in the United States, marked by challenging treatment and dismal prognoses. As immunotherapy emerges as a promising avenue for mitigating PDAC\'s malignant progression, a comprehensive understanding of the tumor\'s immunosuppressive characteristics becomes imperative. This paper systematically delves into the intricate immunosuppressive network within PDAC, spotlighting the significant crosstalk between immunosuppressive cells and factors in the hypoxic acidic pancreatic tumor microenvironment. By elucidating these mechanisms, we aim to provide insights into potential immunotherapy strategies and treatment targets, laying the groundwork for future studies on PDAC immunosuppression. Recognizing the profound impact of immunosuppression on PDAC invasion and metastasis, this discussion aims to catalyze the development of more effective and targeted immunotherapies for PDAC 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
    Pancreatic ductal adenocarcinoma (PDAC) is characterized by poor response to all therapeutic modalities and dismal prognosis. The presence of tertiary lymphoid structures (TLSs) in various solid cancers is of crucial prognostic significance, highlighting the intricate interplay between the tumor microenvironment and immune cells aggregation. However, the extent to which TLS and immune status affect PDAC prognosis remains incompletely understood. Here, we sought to unveil the unique properties of TLS in PDAC by leveraging both single-cell and bulk transcriptomics, and culminating in a risk model that predicts clinical outcomes. We used TLS score based on 12 genes (CCL2, CCL3, CCL4, CCL5, CCL8, CCL18, CCL19, CCL21, CXCL9, CXCL10, CXCL11 and CXCL13) and 9 genes (PTGDS, RBP5, EIF1AY, CETP, SKAP1, LAT, CCR6, CD1D and CD79B) signature, respectively, and examined their distribution in cell clusters of single-cell data from PDAC samples. The markers involved in these clusters were selected to develop a prognostic model using The Cancer Genome Atlas Program (TCGA) database as the training cohort and Gene Expression Omnibus (GEO) database as the validation cohort. Further we compared the immune infiltration, drug sensitivity, enriched and differentially expressed genes between the high-risk and low-risk groups in our model. Therefore, we established a risk model that has significant implications for the prognostic assessment of PADC patients with remarkable differences in immune infiltration and chemo-sensitivity between the low-risk and high-risk groups. And this paradigm established by TLS-related cell marker genes provides a prognostic prediction and a panel of novel therapeutic targets for exploring potential immunotherapy.
    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
    本研究旨在使用从监测中收集的数据比较不同组织学类型的胰腺癌化疗的有效性,流行病学,和结束结果(SEER)数据库。从SEER数据库中选择2004年至2015年间诊断为胰腺癌的患者。采用倾向评分匹配(PSM)来最小化选择偏差。利用Kaplan-Meier存活曲线和对数秩检验来比较不同组之间的总体存活期(OS)和癌症特异性存活期(CSS)。在7,653名胰腺癌患者中,化疗组的OS和CSS均高于非化疗组(p<0.001).PSM之后,产生2381对。Kaplan-Meier生存曲线表明胰腺导管腺癌(PDAC)的OS和CSS,胰腺腺鳞癌(PASC),化疗组胰腺黏液分泌型腺癌(PMPAC)(p<0.001)优于非化疗组,而胰腺黏液腺癌(PMAC)没有显着差异(p=0.2586)。与PASC和PMPAC相比,PDAC展示了更长的OS和CSS。统计学分析结果显示,PASC肿瘤以低分化为主,大多数PMPAC患者有远处转移。化疗可以延长胰腺癌患者的生存期,特别是对于晚期疾病的患者。PMPAC患者有较高的转移率,伴随着更糟糕的生存。
    This study aimed to compare the effectiveness of chemotherapy in different histological types of pancreatic cancer using data collected from the Surveillance, Epidemiology, and End Results (SEER) database. Patients who were diagnosed with pancreatic cancer between 2004 and 2015 were selected from the SEER database. Propensity score matching (PSM) was employed to minimize the selection bias. The Kaplan-Meier survival curves and the log-rank test were utilized to compare the overall survival (OS) and cancer-specific survival (CSS) among different groups. Of the 7,653 pancreatic cancer patients, both OS and CSS were higher in the chemotherapy group than those in the non-chemotherapy group (p < 0.001). After PSM, 2381 pairs were generated. The Kaplan-Meier survival curved indicated that both OS and CSS for pancreatic ductal adenocarcinoma (PDAC), pancreatic adenosquamous carcinoma (PASC), and pancreatic mucin-producing adenocarcinoma (PMPAC) (p < 0.001) in the chemotherapy group were superior to those in the non-chemotherapy group, while there was no significant difference in pancreatic mucinous adenocarcinoma (PMAC) (p = 0.2586). Compared with PASC and PMPAC, PDAC exhibited longer OS and CSS. The results of statistical analysis showed that PASC tumors were mainly poorly differentiated, and the majority of patients with PMPAC had distant metastasis. Chemotherapy could prolong pancreatic cancer patients\' survival, especially for patients with advanced disease. PMPAC patients had a higher rate of metastasis, accompanying with the worse survival.
    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