pancreatic ductal adenocarcinoma

胰腺导管腺癌
  • 文章类型: Journal Article
    目的:本研究的目的是确定特定部位转移模式随时间的作用,并评估与转移性PDAC的延长生存期相关的因素。所有胰腺导管腺癌(PDAC)患者中有一半患有转移性疾病。由于其预后价值,转移部位在临床决策中起着至关重要的作用。
    方法:我们从国家癌症数据库(2016-2019)检查了56,757例IV期PDAC患者,按转移部位分类:多个,肝脏,肺,大脑,骨头,癌,或其他。使用对数秩检验评估特定地点的预后价值,同时通过Aalen的线性风险模型评估随时间变化的影响。用逻辑回归分析评估与延长生存期(>3年)相关的因素。
    结果:仅有远处淋巴结转移(9.0个月)和仅有肺转移(8.1个月)的患者的中位总生存期(mOS)明显长于仅有肝转移(4.6个月,p<0.001)。然而,六个月后,转移部位失去预后价值。Logistic回归确定延长的幸存者(3.6%)更有可能更年轻,西班牙裔,私人保险,Charlson指数<2,接受过化疗,或曾接受过原发性或远处手术(所有p<0.001)。
    结论:虽然同步肝转移比仅肺转移和仅淋巴结转移更差的预后相关,该预测值在6个月后降低.因此,超过此时间的治疗决定不应主要取决于转移部位。在一小部分具有良好肿瘤生物学和良好条件状态的患者中,延长生存期是可能的。他们更有可能接受积极的治疗。
    OBJECTIVE: The aim of this study was to determine the role of site-specific metastatic patterns over time and assess factors associated with extended survival in metastatic PDAC. Half of all patients with pancreatic ductal adenocarcinoma (PDAC) present with metastatic disease. The site of metastasis plays a crucial role in clinical decision making due to its prognostic value.
    METHODS: We examined 56,757 stage-IV PDAC patients from the National Cancer Database (2016-2019), categorizing them by metastatic site: multiple, liver, lung, brain, bone, carcinomatosis, or other. The site-specific prognostic value was assessed using log-rank tests while time-varying effects were assessed by Aalen\'s linear hazards model. Factors associated with extended survival (>3years) were assessed with logistic regression.
    RESULTS: Median overall survival (mOS) in patients with distant lymph node-only metastases (9.0 months) and lung-only metastases (8.1 months) was significantly longer than in patients with liver-only metastases (4.6 months, p < 0.001). However, after six months, the metastatic site lost prognostic value. Logistic regression identified extended survivors (3.6 %) as more likely to be younger, Hispanic, privately insured, Charlson-index <2, having received chemotherapy, or having undergone primary or distant site surgery (all p < 0.001).
    CONCLUSIONS: While synchronous liver metastases are associated with worse outcomes than lung-only and lymph node-only metastases, this predictive value is diminished after six months. Therefore, treatment decisions beyond this time should not primarily depend on the metastatic site. Extended survival is possible in a small subset of patients with favorable tumor biology and good conditional status, who are more likely to undergo aggressive therapies.
    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
    胰腺癌,主要是胰腺导管腺癌(PDAC),仍然是一种高度致命的恶性肿瘤,治疗选择有限,预后不佳。通过靶向导致PDAC发育和进展的潜在分子异常,基因治疗为克服常规放疗和化疗带来的挑战提供了一个有希望的策略.这项研究旨在探索专门针对PDAC中CCAAT/增强子结合蛋白α(CEBPA)基因的小激活RNA(saRNA)的治疗潜力。为了克服与saRNA递送相关的挑战,四面体框架核酸(tFNA)被合理地设计为纳米载体。用截短的运铁蛋白受体适体(tTR14)进一步官能化这些tFNA以增强对PDAC细胞的靶向特异性。构建的基于tFNA的saRNA制剂表现出卓越的稳定性,高效的saRNA释放能力,大量的细胞摄取,生物相容性,和无毒。体外实验显示,利用tTR14修饰的tFNA纳米载体成功地在细胞内递送CEBPA-saRNA,导致抑癌基因的显著激活,即,CEBPA及其下游效应子P21,导致PDAC细胞增殖的显著抑制。此外,在PDAC的小鼠模型中,tTR14修饰的tFNA介导的CEBPA-saRNA的递送有效地上调了CEBPA和P21基因的表达,从而抑制肿瘤生长。这些令人信服的发现强调了通过设计的tFNA纳米载体递送的saRNA作为PDAC的创新治疗方法诱导肿瘤抑制基因激活的潜在效用。
    Pancreatic cancer, predominantly pancreatic ductal adenocarcinoma (PDAC), remains a highly lethal malignancy with limited therapeutic options and a dismal prognosis. By targeting the underlying molecular abnormalities responsible for PDAC development and progression, gene therapy offers a promising strategy to overcome the challenges posed by conventional radiotherapy and chemotherapy. This study sought to explore the therapeutic potential of small activating RNAs (saRNAs) specifically targeting the CCAAT/enhancer-binding protein alpha (CEBPA) gene in PDAC. To overcome the challenges associated with saRNA delivery, tetrahedral framework nucleic acids (tFNAs) were rationally engineered as nanocarriers. These tFNAs were further functionalized with a truncated transferrin receptor aptamer (tTR14) to enhance targeting specificity for PDAC cells. The constructed tFNA-based saRNA formulation demonstrated exceptional stability, efficient saRNA release ability, substantial cellular uptake, biocompatibility, and nontoxicity. In vitro experiments revealed successful intracellular delivery of CEBPA-saRNA utilizing tTR14-decorated tFNA nanocarriers, resulting in significant activation of tumor suppressor genes, namely, CEBPA and its downstream effector P21, leading to notable inhibition of PDAC cell proliferation. Moreover, in a mouse model of PDAC, the tTR14-decorated tFNA-mediated delivery of CEBPA-saRNA effectively upregulated the expression of the CEBPA and P21 genes, consequently suppressing tumor growth. These compelling findings highlight the potential utility of saRNA delivered via a designed tFNA nanocarrier to induce the activation of tumor suppressor genes as an innovative therapeutic approach for PDAC.
    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
    DNA错配修复基因MutL同源物-1(MLH1)在许多癌症中具有不同的作用,然而,其对胰腺导管腺癌(PDAC)转移的影响尚不清楚.在这项研究中,建立了MLH1稳定过表达(OE)和敲除(KD)亚系。伤口愈合和Transwell测定用于评估细胞迁移/侵袭。在原位植入模型(SCID小鼠)中研究体内转移。采用RT-qPCR和蛋白质印迹法显示基因/蛋白质表达。通过转录组测序筛选MLH1下游基因。应用基于组织微阵列的免疫组织化学来确定人样本中的蛋白质表达。在成功生成的子行中,OE细胞表现出较弱的迁移/侵袭能力,与对照组相比,而在KD细胞中,这些能力明显更强。在小鼠中也观察到MLH1的转移抑制作用。机械上,G蛋白偶联受体C5C(GPRC5C)是PDAC细胞MLH1的关键下游基因。随后,瞬时GPRC5C沉默有效抑制细胞迁移/侵袭,并显着逆转了MLH1敲低在KD细胞中的促侵袭作用。在动物模型和人类PDAC组织中,肿瘤GPRC5C表达,与MLH1表达式负相关,与组织学分级呈正相关,船只入侵,癌症特异性生存率低。总之,MLH1通过下调GPRC5C抑制PDAC的转移潜能。
    DNA mismatch repair gene MutL homolog-1 (MLH1) has divergent effects in many cancers, however, its impact on the metastasis of pancreatic ductal adenocarcinoma (PDAC) remains unclear. In this study, MLH1 stably overexpressed (OE) and knockdowned (KD) sub-lines were established. Wound-healing and Transwell assays were used to evaluate cell migration/invasion. In vivo metastasis was investigated in orthotopic implantation models (SCID mice). RT-qPCR and western blotting were adopted to show gene/protein expression. MLH1 down-stream genes were screened by transcriptome sequencing. Tissue microarray-based immunohistochemistry was applied to determine protein expression in human specimens. In successfully generated sub-lines, OE cells presented weaker migration/invasion abilities, compared with controls, while in KD cells these abilities were significantly stronger. The metastasis-inhibitory effect of MLH1 was also observed in mice. Mechanistically, G-protein coupled receptor C5C (GPRC5C) was a key down-stream gene of MLH1 in PDAC cells. Subsequently, transient GPRC5C silencing effectively inhibited cell migration/invasion, and remarkably reversed the pro-invasive effect of MLH1 knockdown in KD cells. In animal models and human PDAC tissues, tumoral GPRC5C expression, negatively associated with MLH1 expressions, was positively correlated with histological grade, vessel invasion, and poor cancer-specific survival. In conclusion, MLH1 inhibits the metastatic potential of PDAC via down-regulation of GPRC5C.
    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
    周围神经系统是癌症进展的关键调节因子。在胰腺导管腺癌(PDAC)中,自主神经系统的交感神经分支抑制癌症的发展。这种抑制作用与早期胰腺癌前体病变中广泛的交感神经发芽有关。然而,这一过程背后的潜在机制仍不清楚.本研究旨在探讨胰腺雪旺细胞在交感神经元结构可塑性中的作用。我们检查了PDAC转基因小鼠模型和慢性炎症诱导的化生胰腺病变模型中雪旺氏细胞数量和分布的变化。在化生/肿瘤性胰腺病变中,雪旺细胞与新的交感神经芽同时增殖和扩张。稀疏遗传标记表明,这些病变中的单个雪旺氏细胞比生理条件下的雪旺氏细胞具有更细长和分支的结构。雪旺细胞过表达神经营养因子,包括胶质细胞源性神经营养因子(GDNF)。交感神经元上调GDNF受体,并在体外响应GDNF表现出增强的神经突生长。雪旺氏细胞中Gdnf的选择性遗传缺失完全阻断了体内化生胰腺病变中交感神经的发芽。这项研究表明,胰腺雪旺细胞在早期癌症发展过程中经历了适应性重编程,支持保护性的抗肿瘤神经元反应。这些发现可能有助于开发调节癌症相关神经可塑性的新策略。
    The peripheral nervous system is a key regulator of cancer progression. In pancreatic ductal adenocarcinoma (PDAC), the sympathetic branch of the autonomic nervous system inhibits cancer development. This inhibition is associated with extensive sympathetic nerve sprouting in early pancreatic cancer precursor lesions. However, the underlying mechanisms behind this process remain unclear. This study aimed to investigate the roles of pancreatic Schwann cells in the structural plasticity of sympathetic neurons. We examined the changes in the number and distribution of Schwann cells in a transgenic mouse model of PDAC and in a model of metaplastic pancreatic lesions induced by chronic inflammation. Schwann cells proliferated and expanded simultaneously with new sympathetic nerve sprouts in metaplastic/neoplastic pancreatic lesions. Sparse genetic labeling showed that individual Schwann cells in these lesions had a more elongated and branched structure than those under physiological conditions. Schwann cells overexpressed neurotrophic factors, including glial cell-derived neurotrophic factor (GDNF). Sympathetic neurons upregulated the GDNF receptors and exhibited enhanced neurite growth in response to GDNF in vitro. Selective genetic deletion of Gdnf in Schwann cells completely blocked sympathetic nerve sprouting in metaplastic pancreatic lesions in vivo. This study demonstrated that pancreatic Schwann cells underwent adaptive reprogramming during early cancer development, supporting a protective antitumor neuronal response. These finding could help to develop new strategies to modulate cancer associated neural plasticity.
    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
    带有淋巴结(LN)解剖的胰体远端切除术(DP)是胰腺尾部导管腺癌(Pt-PDAC)的标准程序。然而,包括LN夹层范围在内的最佳手术仍在争论中。本研究调查了LN转移对患有Pt-PDAC的患者的发生率和预后影响。
    这个多中心,回顾性研究纳入了2013年至2017年间在12个机构接受了可切除Pt-PDACDP治疗的163例患者.研究了LN转移的频率以及LN解剖对Pt-PDAC预后的影响。
    在患有Pt-PDAC的患者中,沿着脾动脉的LN转移的发生率很高(39%)。LN沿共同肝的转移率,左胃,腹腔动脉很低,这些LN的治疗指数为零。在位于远端的胰腺尾癌中,LN沿肝总动脉无转移.多因素分析显示肿瘤大小是影响无复发生存率的独立因素(HR=2.01,95%CI=1.33~3.05,p=0.001)。沿着肝总动脉的胰腺分裂和LN解剖水平不影响肿瘤复发或无复发生存的部位。
    对Pt-PDAC沿肝动脉进行LN解剖意义不大。就肿瘤安全性而言,远端胰腺横切术可能是可以接受的,但需要进一步检查短期结局和胰腺功能的保留.
    UNASSIGNED: Distal pancreatectomy (DP) with lymph node (LN) dissection is the standard procedure for pancreatic ductal adenocarcinoma of the tail (Pt-PDAC). However, the optimal surgery including extent of LN dissection is still being debated. The present study investigated the incidence and prognostic impact of LN metastasis on patients suffering from Pt-PDAC.
    UNASSIGNED: This multicenter, retrospective study involved 163 patients who underwent DP for resectable Pt-PDAC at 12 institutions between 2013 and 2017. The frequency of LN metastasis and the effect of LN dissection on Pt-PDAC prognosis were investigated.
    UNASSIGNED: There were high incidences of metastases to the LNs along the splenic artery in the patients with Pt-PDAC (39%). The rate of metastases in the LNs along the common hepatic, left gastric, and celiac arteries were low, and the therapeutic index for these LNs was zero. In pancreatic tail cancer located more distally, there were no metastases to the LNs along the common hepatic artery. Multivariate analysis revealed that tumor size was the only independent factor related to recurrence-free survival (HR = 2.01, 95% CI = 1.33-3.05, p = 0.001). The level of pancreas division and LN dissection along the common hepatic artery did not affect the site of tumor recurrence or recurrence-free survival.
    UNASSIGNED: LN dissection along the hepatic artery for Pt-PDAC has little significance. Distal pancreatic transection may be acceptable in terms of oncological safety, but further examination of short-term outcomes and preservation of pancreatic function is 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
    背景:胰腺导管腺癌(PDAC)患者口腔改变,胃肠,和胰腺内微生物组与健康个体相比。然而,关于胆汁微生物组及其对PDAC无进展生存期的潜在影响的知识仍然有限.
    方法:PDAC患者(n=45),包括手术前后的20对配对,和良性对照(n=16)被纳入前瞻性研究。通过16S-rRNA基因测序揭示了总共81个胆汁的微生物群落特征。PDAC患者根据肿瘤标志物水平分为不同的组,疾病分期,手术前后,以及无进展生存期(PFS)进行进一步分析。利用随机森林算法建立疾病诊断模型。
    结果:PDAC患者拥有独特多样的胆汁微生物组(PCoA,加权Unifrac,p=0.038),根据关键微生物和微生物功能,微生物多样性的增加与菌群失调相关。Aliihoeflea是两组中表现出最显着变化的属(p<0.01)。发现长期PFS和短期PFS组之间胆汁微生物组的β多样性存在显着差异(PCoA,加权Unifrac,p=0.005)。在所有PDAC患者中,杆菌和放线菌被确定为与无进展生存期相关的两组之间的门改变。此外,我们确定了三个生物标志物作为随机森林模型的最合适的集合,这表明PDAC组发生疾病的可能性显着升高(p<0.0001)。受试者工作特征(ROC)曲线下面积达到80.8%,95%置信区间为55.0%至100%。由于胆汁样本的稀缺性,我们无法进行进一步的外部核查。
    结论:PDAC的特征是胆管微生物组改变。胆道菌群失调与所有PDAC的无进展生存期相关。这项研究揭示了PDAC中胆汁微生物组的改变,并成功开发了PDAC的诊断模型。
    BACKGROUND: Patients with pancreatic ductal adenocarcinoma (PDAC) display an altered oral, gastrointestinal, and intra-pancreatic microbiome compared to healthy individuals. However, knowledge regarding the bile microbiome and its potential impact on progression-free survival in PDACs remains limited.
    METHODS: Patients with PDAC (n = 45), including 20 matched pairs before and after surgery, and benign controls (n = 16) were included prospectively. The characteristics of the microbiomes of the total 81 bile were revealed by 16  S-rRNA gene sequencing. PDAC patients were divided into distinct groups based on tumor marker levels, disease staging, before and after surgery, as well as progression free survival (PFS) for further analysis. Disease diagnostic model was formulated utilizing the random forest algorithm.
    RESULTS: PDAC patients harbor a unique and diverse bile microbiome (PCoA, weighted Unifrac, p = 0.038), and the increasing microbial diversity is correlated with dysbiosis according to key microbes and microbial functions. Aliihoeflea emerged as the genus displaying the most significant alteration among two groups (p < 0.01). Significant differences were found in beta diversity of the bile microbiome between long-term PFS and short-term PFS groups (PCoA, weighted Unifrac, p = 0.005). Bacillota and Actinomycetota were identified as altered phylum between two groups associated with progression-free survival in all PDAC patients. Additionally, we identified three biomarkers as the most suitable set for the random forest model, which indicated a significantly elevated likelihood of disease occurrence in the PDAC group (p < 0.0001). The area under the receiver operating characteristic (ROC) curve reached 80.8% with a 95% confidence interval ranging from 55.0 to 100%. Due to the scarcity of bile samples, we were unable to conduct further external verification.
    CONCLUSIONS: PDAC is characterized by an altered microbiome of bile ducts. Biliary dysbiosis is linked with progression-free survival in all PDACs. This study revealed the alteration of the bile microbiome in PDACs and successfully developed a diagnostic model for 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
    Objective To construct a risk prediction model by integrating the molecular subtypes of pancreatic ductal adenocarcinoma (PDAC) and immune-related genes.Methods With GSE71729 data set (n=145) as the training set,the differentially expressed genes and differential immune-related genes between the squamous and non-squamous subtypes of PDAC were integrated to construct a regulatory network,on the basis of which five immune marker genes regulating the squamous subtype were screened out.An integrated immune score (IIS) model was constructed based on patient survival information and immune marker genes to predict the clinical prognosis of PDAC patients,and its predictive performance was tested with 5 validation sets (n=758).Results PDAC patients were assigned into high risk and low risk groups according to the IIS.In both training and validation sets,the overall survival of patients in the high risk group was shorter than that in the low risk group (both P<0.001).The multivariable Cox regression showed that IIS was an independent prognostic factor for PDAC (HR=2.16,95%CI=1.50-3.10,P<0.001).Conclusion IIS can be used for risk stratification of PDAC patients and may become a potential prognostic marker for PDAC.
    目的 构建整合胰腺导管腺癌(PDAC)分子亚型和免疫相关基因的风险评估模型。方法 以GSE71729数据集(n=145)为训练集,整合PDAC鳞状和非鳞状亚型之间差异表达基因和差异免疫相关基因构建调控网络,筛选发挥主调控鳞状亚型的5个免疫标志基因。基于患者生存信息和免疫标志基因构建整合免疫评分(IIS)模型来预测PDAC患者的临床预后,并在5个验证集(n=758)中检验其预测效能。结果 根据IIS将PDAC患者分为高危组和低危组。在训练集和验证集中,高危组患者的总体生存期均短于低危组(P均<0.001)。多变量Cox回归分析显示IIS是PDAC的独立预后因子(HR=2.16,95%CI=1.50~3.10,P<0.001)。结论 IIS可用于PDAC患者的危险分层,并可能成为PDAC潜在的预后标志物。.
    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
    NK细胞是胰腺导管腺癌(PDAC)肿瘤微环境的重要组成部分,在PDAC开发中也发挥着重要作用。本研究旨在探讨NK细胞标记基因与预后的关系,PDAC患者的免疫反应。根据scRNA-seq数据,我们发现NK细胞的比例在PDAC中显著下调,筛选出373个NK细胞标记基因。通过TCGA数据库,我们纳入了7个NK细胞标记基因,以构建预测PDAC患者预后的标记.Cox分析确定该特征是胰腺癌的独立因素。随后,通过6个GEO数据集验证了特征的预测能力,并获得了出色的评价.我们对签名与患者免疫状态之间的关系的分析显示,签名与免疫细胞浸润有很强的相关性,炎症反应,免疫检查点抑制剂(ICIs)反应。NK细胞标记基因与PDAC患者的预后和免疫功能密切相关,它们具有作为治疗靶点的潜在价值。
    The NK cell is an important component of the tumor microenvironment of pancreatic ductal adenocarcinoma (PDAC), also plays a significant role in PDAC development. This study aimed to explore the relationship between NK cell marker genes and prognosis, immune response of PDAC patients. By scRNA-seq data, we found the proportion of NK cells were significantly downregulated in PDAC and 373 NK cell marker genes were screened out. By TCGA database, we enrolled 7 NK cell marker genes to construct the signature for predicting prognosis in PDAC patients. Cox analysis identified the signature as an independent factor for pancreatic cancer. Subsequently, the predictive power of signature was validated by 6 GEO datasets and had an excellent evaluation. Our analysis of relationship between the signature and patients\' immune status revealed that the signature has a strong correlation with immunocyte infiltration, inflammatory reaction, immune checkpoint inhibitors (ICIs) response. The NK cell marker genes are closely related to the prognosis and immune capacity of PDAC patients, and they have potential value as a therapeutic target.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: English Abstract
    探讨胰腺导管腺癌(PDAC)根治性切除术患者的基线临床特征和血液学参数与预后的关系。并为患者临床风险分层提供参考。
    我们回顾性收集了在华西医院接受PDAC根治性手术治疗的445例患者的临床资料,四川大学2010年1月至2019年2月。然后,我们对收集的数据进行了回顾性临床分析.关于患者基本临床特征的数据,血常规检查结果,收集肿瘤指标,探讨其对PDAC患者术后总生存期(OS)的影响。Cox比例风险回归用于确定影响OS的因素。采用SPSS23.0软件包进行统计学分析。
    术后中位总生存期(mOS)为17.0个月(95%CI:15.0-19.0)。纳入研究的患者的1、2、3、4和5年生存率为60.6%,33.4%,19.1%,12.7%,9.6%,分别。多变量Cox比例风险模型分析表明,许多因素独立影响PDAC患者的术后生存率。这些因素包括肿瘤位置(危险比[HR]=1.574,95%CI:1.233-2.011),肿瘤细胞分化程度(HR=0.687,95%CI:0.542-0.870),存在神经浸润(HR=0.686,95%CI:0.538-0.876),TNM分期(HR=1.572,95%CI:1.252-1.974),术后辅助治疗(HR=1.799,95%CI:1.390-2.328),术前饮酒史(HR=0.744,95%CI:0.588-0.943),术前血清CA199水平较高(HR=0.742,95%CI:0.563-0.977)。
    在PDAC患者中,胰腺头部有肿瘤,中度和高度分化,没有局部神经血管侵犯,处于TNM第一阶段,接受术后辅助治疗,手术前没有饮酒史,术前血清CA199小于或等于37U/mL与更好的预后显著相关。
    UNASSIGNED: To explore the relationship between baseline clinical characteristics and hematological parameters of patients undergoing radical resection for pancreatic ductal adenocarcinoma (PDAC) and their prognosis, and to provide references for stratifying the patients\' clinical risks.
    UNASSIGNED: We retrospectively collected clinical data from 445 patients who underwent radical surgical treatment for PDAC at West China Hospital, Sichuan University between January 2010 and February 2019. Then, we conducted retrospective clinical analysis with the collected data. Data on patients\' basic clinical characteristics, routine blood test results, and tumor indicators were collected to explore their effects on the postoperative overall survival (OS) of PDAC patients. Cox proportional hazards regression was used to identify factors affecting OS. Statistical analysis was performed using the SPSS 23.0 software package.
    UNASSIGNED: The postoperative median overall survival (mOS) was 17.0 months (95% CI: 15.0-19.0). The 1, 2, 3, 4, and 5-year survival rates of the patients included in the study were 60.6%, 33.4%, 19.1%, 12.7%, and 9.6%, respectively. The multivariate Cox proportional hazards model analysis demonstrated that a number of factors independently affect postoperative survival in PDAC patients. These factors include tumor location (hazards ratio [HR]=1.574, 95% CI: 1.233-2.011), degree of tumor cell differentiation (HR=0.687, 95% CI: 0.542-0.870), presence of neural invasion (HR=0.686, 95% CI: 0.538-0.876), TNM staging (HR=1.572, 95% CI: 1.252-1.974), postoperative adjuvant therapy (HR=1.799, 95% CI: 1.390-2.328), preoperative drinking history (HR=0.744, 95% CI: 0.588-0.943), and high serum CA199 levels prior to the surgery (HR=0.742, 95% CI: 0.563-0.977).
    UNASSIGNED: In PDAC patients, having tumors located in the head of the pancreas, moderate and high degrees of differentiated, being free from local neurovascular invasion, being in TNM stage Ⅰ, undergoing postoperative adjuvant therapy, no history of alcohol consumption prior to the surgery, and preoperative serum CA199 being less than or equal to 37 U/mL are significantly associated with a better prognosis.
    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)迫切需要二线或后期治疗策略。我们旨在分析额外安洛替尼的疗效和安全性,特别是安洛替尼与免疫治疗联合使用,一线治疗失败的PDAC患者。
    病理诊断为PDAC的患者接受安洛替尼治疗,一些患者同时接受抗PD-1药物治疗,可以进行回顾性分析。评估额外安洛替尼的疗效和安全性。
    共纳入23例患者。在接受额外安洛替尼治疗的患者中,中位无进展生存期(PFS)为1.8个月,中位总生存期(OS)为6.3个月,无论抗PD-1药物。在接受额外安洛替尼与抗PD-1药物联合治疗的患者中,中位PFS和OS分别为1.8和6.5个月,分别。16例患者(69.6%)发生不良事件。在接受额外安洛替尼治疗的患者中,大多数AE为1-3级。单变量分析显示,基线红细胞分布宽度(RDW)<14%的患者接受额外的安洛替尼联合抗PD-1药物治疗,其OS明显长于基线RDW≥14%的患者(p=0.025)。使用额外的安洛替尼联合抗PD-1药物作为二线治疗的患者比作为后期治疗的患者具有更长的OS(p=0.012)。多因素分析显示,基线RDW是OS的唯一独立危险因素(p=0.042)。
    安洛替尼和免疫疗法的组合是PDAC患者的一种有效的附加疗法,具有可耐受的不良事件,可作为二线或后期治疗,特别是在基线RDW<14%的患者中。
    UNASSIGNED: Pancreatic ductal adenocarcinoma (PDAC) is in urgent need of a second-line or later-line treatment strategy. We aimed to analyze the efficacy and safety of additional anlotinib, specifically anlotinib in combination with immunotherapy, in patients with PDAC who have failed first-line therapy.
    UNASSIGNED: Patients with pathological diagnosis of PDAC were additionally treated with anlotinib, and some patients were treated with anti-PD-1 agents at the same time, which could be retrospectively analyzed. The efficacy and safety of additional anlotinib were evaluated.
    UNASSIGNED: A total of 23 patients were included. In patients treated with additional anlotinib, the overall median progression-free survival (PFS) was 1.8 months and the median overall survival (OS) was 6.3 months, regardless of anti-PD-1 agents. Among patients receiving additional anlotinib in combination with anti-PD-1 agents, median PFS and OS were 1.8 and 6.5 months, respectively. Adverse events (AEs) were observed in 16 patients (69.6%). In patients treated with additional anlotinib, the majority of AEs were grade 1-3. Univariate analysis revealed that patients with baseline red blood cell distribution width (RDW) <14% treated with additional anlotinib plus anti-PD-1 agents had significantly longer OS than patients with baseline RDW ≥14% (p = 0.025). Patients with additional anlotinib plus anti-PD-1 agents as second-line therapy had a longer OS than those treated as later-line therapy (p = 0.012). Multivariate analysis showed that baseline RDW was the only independent risk factor for OS (p = 0.042).
    UNASSIGNED: The combination of anlotinib and immunotherapy represents an effective add-on therapy with tolerable AEs as second- or later-line therapy in patients with PDAC, particularly in patients with baseline RDW <14%.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号