Deoxycytidine

脱氧胞苷
  • 文章类型: Case Reports
    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
    耐药性仍然是胰腺癌治疗中的重大挑战。耐药细胞系的开发对于理解耐药的潜在机制和开发新药以改善临床结果至关重要。这里,一种新的胰腺癌细胞系,来自中国患者的PDAC-X1已经建立。PDAC-X1的特点是免疫表型,生物学遗传学,分子特征,和致瘤性。体外分析显示,PDAC-X1细胞表现出上皮形态和细胞标志物(CK7和CK19),表达的癌症相关标志物(E-钙黏着蛋白,Vimentin,Ki-67,CEA,CA19-9),并在悬浮培养中产生胰腺癌样器官。体内分析显示PDAC-X1细胞以100%的肿瘤形成率维持致瘤性。该细胞系表现出复杂的核型,以亚三倍体核型为主。此外,PDAC-X1细胞对多种药物表现出内在的多药耐药性,包括吉西他滨,紫杉醇,5-氟尿嘧啶,和奥沙利铂.总之,PDAC-X1细胞系已经建立和表征,代表一个有用的和有价值的临床前模型来研究潜在的耐药机制,并开发新的药物疗法,以改善患者的结果。
    Drug resistance remains a significant challenge in the treatment of pancreatic cancer. The development of drug-resistant cell lines is crucial to understanding the underlying mechanisms of resistance and developing novel drugs to improve clinical outcomes. Here, a novel pancreatic cancer cell line, PDAC-X1, derived from Chinese patients has been established. PDAC-X1 was characterized by the immune phenotype, biology, genetics, molecular characteristics, and tumorigenicity. In vitro analysis revealed that PDAC-X1 cells exhibited epithelial morphology and cell markers (CK7 and CK19), expressed cancer-associated markers (E-cadherin, Vimentin, Ki-67, CEA, CA19-9), and produced pancreatic cancer-like organs in suspension culture. In vivo analysis showed that PDAC-X1 cells maintained tumorigenicity with a 100% tumor formation rate. This cell line exhibited a complex karyotype, dominated by subtriploid karyotypes. In addition, PDAC-X1 cells exhibited intrinsic multidrug resistance to multiple drugs, including gemcitabine, paclitaxel, 5-fluorouracil, and oxaliplatin. In conclusion, the PDAC-X1 cell line has been established and characterized, representing a useful and valuable preclinical model to study the underlying mechanisms of drug resistance and develop novel drug therapeutics to improve patient outcomes.
    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
    吉西他滨(GEM)耐药性的发展严重限制了胰腺癌(PC)的治疗功效,越来越多的证据强调了环状RNA(circularRNAs)在肿瘤发生中的重要作用。PC的进展和耐药性。然而,PC的GEM抗性发展的潜在circRNAs仍有待澄清。当前的研究旨在揭示circ_0036627在指示PC中的侵略性和GEM敏感性中的作用。我们报道了circ_0036627在PC组织和PC细胞系中的表达增加。升高的circ_0036627表达水平与PC患者的晚期肿瘤分级和较差的总体生存率相关。功能测定和体内实验表明circ_0036627过表达是增殖所必需的,PC细胞的迁移侵袭和GEM抗性。circ_0036627敲低抑制了体内肿瘤的发展。分子分析进一步显示circ_0036627通过海绵microRNA-145(miR-145)增加S100A16的表达,一种可以显著减弱PC细胞增殖的肿瘤抑制性miRNA,迁移,入侵和GEM抵抗。此外,我们的研究结果表明,S100A16作为致癌因子促进PC细胞的侵袭性和GEM抗性。总之,当前的发现为PC侵略性和GEM抗性提供了新的机械见解,提示circ_0036627/miR-145/S100A16轴在PC进展和耐药性发展中的关键作用,并为PC治疗提供新的治疗靶标。
    The development of gemcitabine (GEM) resistance severely limits the treatment efficacy in pancreatic cancer (PC) and increasing evidence highlights the vital roles of circular RNAs (circRNAs) in the tumorigenesis, progression and drug resistance of PC. However, the circRNAs underlying GEM resistance development of PC remains to be clarified. The current research aims to unveil the roles of circ_0036627 in dictating the aggressiveness and GEM sensitivity in PC. We reported the increased expression of circ_0036627 in PC tissues and PC cell lines. Elevated circ_0036627 expression level was correlated with advanced tumour grade and poor overall survival in PC patients. Functional assays and in vivo experiments demonstrated that circ_0036627 overexpression was required for the proliferation, migration invasion and GEM resistance in PC cells. circ_0036627 knockdown suppressed tumour development in vivo. The molecular analysis further showed that circ_0036627 increased S100A16 expression by sponging microRNA-145 (miR-145), a tumour-suppressive miRNA that could significantly attenuate PC cell proliferation, migration, invasion and GEM resistance. Furthermore, our findings suggested that S100A16 acted as an oncogenic factor to promote aggressiveness and GEM resistance in PC cells. In conclusion, the current findings provide new mechanistic insights into PC aggressiveness and GEM resistance, suggesting the critical role of circ_0036627/miR-145/S100A16 axis in PC progression and drug resistance development and offering novel therapeutic targets for PC therapy.
    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:手术切除后的癌症复发是治疗失败的主要原因。寻找预防术后复发和切口感染的有效方法是手术成功的重要组成部分。随着新纳米技术的发展,为术后辅助治疗提供了更多的治疗选择.这项研究提出了一种创新的水凝胶系统,可以刺激非小细胞肺癌(NSCLC)手术切除后的杀肿瘤免疫并防止癌症复发。
    结果:水凝胶系统基于单原子铂(CN-Pt)的出色光热转化性能以及化疗药物的递送和释放,吉西他滨(GEM)。在肿瘤切除后,通过随后的近红外(NIR)光热疗法将系统涂覆到伤口表面,有效诱导残余癌细胞坏死,放大损伤相关分子模式(DAMP)的水平,并增加M1巨噬细胞的数量。显著较高水平的吞噬巨噬细胞增强肿瘤的免疫原性,并使癌细胞对CD8+T细胞免疫敏感,以控制术后复发。这已经用肺癌术后复发的动物模型进行了验证。具有NIR的CN-Pt-GEM-水凝胶还可以抑制术后伤口感染。
    结论:这些发现为接受NSCLC肿瘤切除的患者提供了一种补充抗肿瘤免疫的替代策略。具有NIR系统的CN-Pt-GEM-水凝胶还表现出良好的生物安全性,并且可能适用于与肿瘤切除手术有关的临床应用。伤口组织填充,预防感染,预防复发。
    BACKGROUND: Cancer recurrence following surgical resection is a major cause of treatment failure. Finding effective methods to prevent postoperative recurrence and wound infection is an important component of successful surgery. With the development of new nanotechnology, more treatment options have been provided for postoperative adjuvant therapy. This study presents an innovative hydrogel system that stimulates tumoricidal immunity after surgical resection of non-small cell lung cancer (NSCLC) and prevents cancer relapse.
    RESULTS: The hydrogel system is based on the excellent photothermal conversion performance of single-atom platinum (CN-Pt) along with the delivery and release of the chemotherapy drug, gemcitabine (GEM). The system is coated onto the wound surface after tumor removal with subsequent near-infrared (NIR) photothermal therapy, which efficiently induces necroptosis of residual cancer cells, amplifies the levels of damage-associated molecular patterns (DAMPs), and increases the number of M1 macrophages. The significantly higher levels of phagocytic macrophages enhance tumor immunogenicity and sensitize cancer cells to CD8 + T-cell immunity to control postoperative recurrence, which has been verified using an animal model of postoperative lung cancer recurrence. The CN-Pt-GEM-hydrogel with NIR can also inhibit postoperative wound infection.
    CONCLUSIONS: These findings introduce an alternative strategy for supplementing antitumor immunity in patients undergoing resection of NSCLC tumors. The CN-Pt-GEM-hydrogel with the NIR system also exhibits good biosafety and may be adaptable for clinical application in relation to tumor resection surgery, wound tissue filling, infection prevention, and recurrence prevention.
    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
    目的:比较nab-紫杉醇的有效性和安全性,顺铂,和卡培他滨(nab-TPC)联合吉西他滨和顺铂作为复发或转移性鼻咽癌的替代一线治疗选择。
    方法:阶段3,开放标签,多中心,随机试验。
    方法:2019年9月至2022年8月,中国有四家医院。
    方法:患有复发性或转移性鼻咽癌的成人(≥18岁)。
    方法:患者以1:1的比例随机分配给nab-紫杉醇(第1天200g/m2),顺铂(第1天60mg/m2),和卡培他滨(第1-14天两次1000mg/m2)或吉西他滨(第1天和第8天1g/m2)和顺铂(第1天80mg/m2)。
    方法:由独立审查委员会评估无进展生存期作为意向治疗人群的主要终点。
    结果:在预设的中期分析中,中位随访时间为15.8个月(2022年10月31日)。根据独立审查委员会的评估,nab-TPC队列的中位无进展生存期为11.3个月(95%置信区间9.7~12.9个月),而吉西他滨和顺铂队列的中位无进展生存期为7.7个月(6.5~9.0个月).风险比为0.43(95%置信区间为0.25至0.73;P=0.002)。nab-TPC组的客观缓解率为83%(34/41),而吉西他滨和顺铂组的客观缓解率为63%(25/40)(P=0.05),nab-TPC队列的缓解持续时间为10.8个月,而吉西他滨和顺铂队列的缓解持续时间为6.9个月(P=0.009).治疗相关的3级或4级不良事件,包括白细胞减少症(4/41(10%)v13/40(33%);P=0.02),中性粒细胞减少症(6/41(15%)v16/40(40%);P=0.01),贫血(1/41(2%)v8/40(20%);P=0.01),吉西他滨和顺铂队列高于nab-TPC队列。在任一治疗组中均未发生与治疗相关的死亡。生存和长期毒性仍在进行长期随访评估。
    结论:与吉西他滨和顺铂相比,nab-TPC方案对复发或转移性鼻咽癌具有更高的抗肿瘤疗效和良好的安全性。Nab-TPC应被视为复发性或转移性鼻咽癌的标准一线治疗方法。需要更长时间的随访以确认总体生存的益处。
    背景:中国临床试验注册ChiCTR1900027112.
    To compare the effectiveness and safety of nab-paclitaxel, cisplatin, and capecitabine (nab-TPC) with gemcitabine and cisplatin as an alternative first line treatment option for recurrent or metastatic nasopharyngeal carcinoma.
    Phase 3, open label, multicentre, randomised trial.
    Four hospitals located in China between September 2019 and August 2022.
    Adults (≥18 years) with recurrent or metastatic nasopharyngeal carcinoma.
    Patients were randomised in a 1:1 ratio to treatment with either nab-paclitaxel (200 g/m2 on day 1), cisplatin (60 mg/m2 on day 1), and capecitabine (1000 mg/m2 twice on days 1-14) or gemcitabine (1 g/m2 on days 1 and 8) and cisplatin (80 mg/m2 on day 1).
    Progression-free survival was evaluated by the independent review committee as the primary endpoint in the intention-to-treat population.
    The median follow-up was 15.8 months in the prespecified interim analysis (31 October 2022). As assessed by the independent review committee, the median progression-free survival was 11.3 (95% confidence interval 9.7 to 12.9) months in the nab-TPC cohort compared with 7.7 (6.5 to 9.0) months in the gemcitabine and cisplatin cohort. The hazard ratio was 0.43 (95% confidence interval 0.25 to 0.73; P=0.002). The objective response rate in the nab-TPC cohort was 83% (34/41) versus 63% (25/40) in the gemcitabine and cisplatin cohort (P=0.05), and the duration of response was 10.8 months in the nab-TPC cohort compared with 6.9 months in the gemcitabine and cisplatin cohort (P=0.009). Treatment related grade 3 or 4 adverse events, including leukopenia (4/41 (10%) v 13/40 (33%); P=0.02), neutropenia (6/41 (15%) v 16/40 (40%); P=0.01), and anaemia (1/41 (2%) v 8/40 (20%); P=0.01), were higher in the gemcitabine and cisplatin cohort than in the nab-TPC cohort. No deaths related to treatment occurred in either treatment group. Survival and long term toxicity are still being evaluated with longer follow-up.
    The nab-TPC regimen showed a superior antitumoural efficacy and favourable safety profile compared with gemcitabine and cisplatin for recurrent or metastatic nasopharyngeal carcinoma. Nab-TPC should be considered the standard first line treatment for recurrent or metastatic nasopharyngeal carcinoma. Longer follow-up is needed to confirm the benefits for overall survival.
    Chinese Clinical Trial Registry ChiCTR1900027112.
    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
    胰腺癌是恶性肿瘤相关死亡的第四大原因,随着胰腺癌干细胞驱动的耐药性的迅速发展。然而,胰腺导管腺癌(PDAC)的干性和化疗耐药维持机制尚不清楚.这里,我们证明了双尾C同源物1(BICC1),调节大量细胞质mRNA的RNA结合蛋白,促进PDAC中的化学抗性和干性。机械上,BICC1通过上调吲哚胺2,3-双加氧酶-1(IDO1)表达激活PDAC中的色氨酸分解代谢,一种色氨酸分解代谢酶.色氨酸代谢物水平的增加有助于NAD+合成和氧化磷酸化,导致干细胞样表型。阻断BICC1/IDO1/色氨酸代谢信号传导极大地改善了具有高BICC1水平的若干PDAC模型中的吉西他滨(GEM)功效。这些发现表明BICC1是驱动PDAC的干性和化学抗性的关键色氨酸代谢调节剂,因此是针对化学抗性的组合治疗策略的潜在靶标。
    Pancreatic adenocarcinoma is the fourth leading cause of malignancy-related deaths, with rapid development of drug resistance driven by pancreatic cancer stem cells. However, the mechanisms sustaining stemness and chemotherapy resistance in pancreatic ductal adenocarcinoma (PDAC) remain unclear. Here, we demonstrate that Bicaudal C homolog 1 (BICC1), an RNA binding protein regulating numerous cytoplasmic mRNAs, facilitates chemoresistance and stemness in PDAC. Mechanistically, BICC1 activated tryptophan catabolism in PDAC by up-regulating indoleamine 2,3-dioxygenase-1 (IDO1) expression, a tryptophan-catabolizing enzyme. Increased levels of tryptophan metabolites contribute to NAD+ synthesis and oxidative phosphorylation, leading to a stem cell-like phenotype. Blocking BICC1/IDO1/tryptophan metabolism signaling greatly improves the gemcitabine (GEM) efficacy in several PDAC models with high BICC1 level. These findings indicate that BICC1 is a critical tryptophan metabolism regulator that drives the stemness and chemoresistance of PDAC and thus a potential target for combinatorial therapeutic strategy against chemoresistance.
    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
    目的:我们试图研究在吉西他滨基础上加用尼妥珠单抗是否能提高晚期胰腺癌的疗效。
    方法:本回顾性分析共纳入98例晚期胰腺癌住院患者。根据具体的治疗方法,将患者分为研究组和对照组。临床疗效,不良反应,并对2组的随访结果进行比较,和身体状况,监测并记录治疗前后的CA724、CA19-9和CEA水平。
    结果:治疗后,PR比率,SD比率,ORR,研究组DCR明显高于对照组,治疗后研究组KPS评分明显高于对照组(P<0.05)。治疗后,然而,3项指标均显著低于对照组(P<0.05)。
    结论:我们的研究强调尼妥珠单抗和吉西他滨的联合疗效优于对照方案,晚期胰腺癌患者的生存率提高,CA724,CA19-9和CEA水平降低。
    OBJECTIVE: We sought to investigate whether the addition of nimotuzumab to gemcitabine would improve the treatment efficacy of advanced pancreatic cancer.
    METHODS: This retrospective analysis involved a total of 98 hospitalized patients harboring advanced pancreatic cancer. Depending on the specific treatment, patients were divided into study groups and control groups. The clinical efficacy, adverse reactions, and follow-up results of the 2 groups were compared, and the physical status, CA724, CA19-9, and CEA levels before and after treatment were monitored and recorded.
    RESULTS: After treatment, PR ratio, SD ratio, ORR, and DCR in the study group were significantly higher than those in the control group, and PD ratio was significantly lower than that in the control group (P < 0.05) the KPS score after treatment in the study group was markedly higher than that of the control group (P < 0.05). After treatment, however, significantly lower levels of the 3 indicators were observed when compared with the control group (P < 0.05).
    CONCLUSIONS: Our study highlights a more superior combined efficacy of nimotuzumab and gemcitabine than the control regimen, exhibiting improved survival and reduced levels of CA724, CA19-9, and CEA in patients with advanced pancreatic cancer.
    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
    胰腺导管腺癌(PDAC)/胰腺癌,是一种高度侵袭性的恶性肿瘤,预后差。基于吉西他滨的化疗仍然是PDAC治疗的基石。尽管如此,患者对吉西他滨耐药的发展是导致预后不良的主要因素.肿瘤表现出的耐药性受一系列因素的调节,如基因突变,肿瘤微环境转变,环境污染物暴露。目前,对环境污染物与肿瘤耐药性之间关系的理解仍然不足。我们的研究发现,PFOS/6:2Cl-PFESA暴露会增加PDAC对吉西他滨的耐药性。随后的体内试验证实,暴露于PFOS/6:2Cl-PFESA可降低吉西他滨抑制PDAC的功效,抑制率从79.5%下降到56.7%/38.7%,分别。整合的多组学测序和分子生物学分析已确定核糖核苷酸还原酶催化亚基M1(RRM1)的上调是吉西他滨耐药的关键因素。随后的研究表明,暴露于全氟辛烷磺酸和6:2Cl-PFESA会导致RRM1途径上调,从而增强化疗抗性。值得注意的是,6:2Cl-PFESA的影响超过了PFOS。尽管6:2Cl-PFESA被认为是全氟辛烷磺酸的更安全替代品,其对PDAC化疗耐药的显著影响需要对其与胃肠道毒性相关的潜在风险进行全面评估.
    Pancreatic ductal adenocarcinoma (PDAC)/pancreatic cancer, is a highly aggressive malignancy with poor prognosis. Gemcitabine-based chemotherapy remains the cornerstone of PDAC treatment. Nonetheless, the development of resistance to gemcitabine among patients is a major factor contributing to unfavorable prognostic outcomes. The resistance exhibited by tumors is modulated by a constellation of factors such as genetic mutations, tumor microenvironment transforms, environmental contaminants exposure. Currently, comprehension of the relationship between environmental pollutants and tumor drug resistance remains inadequate. Our study found that PFOS/6:2 Cl-PFESA exposure increases resistance to gemcitabine in PDAC. Subsequent in vivo trials confirmed that exposure to PFOS/6:2 Cl-PFESA reduces gemcitabine\'s efficacy in suppressing PDAC, with the inhibition rate decreasing from 79.5 % to 56.7 %/38.7 %, respectively. Integrative multi-omics sequencing and molecular biology analyses have identified the upregulation of ribonucleotide reductase catalytic subunit M1 (RRM1) as a critical factor in gemcitabine resistance. Subsequent research has demonstrated that exposure to PFOS and 6:2 Cl-PFESA results in the upregulation of the RRM1 pathway, consequently enhancing chemotherapy resistance. Remarkably, the influence exerted by 6:2 Cl-PFESA exceeds that of PFOS. Despite 6:2 Cl-PFESA being regarded as a safer substitute for PFOS, its pronounced effect on chemotherapeutic resistance in PDAC necessitates a thorough evaluation of its potential risks related to gastrointestinal toxicity.
    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
    骨髓来源的抑制细胞(MDSC)在促进肿瘤免疫逃逸和诱导免疫抑制肿瘤微环境中发挥了重要作用。消除MDSCs和肿瘤细胞仍然是癌症免疫治疗的主要挑战。使用吉西他滨-塞来昔布双药物纳米组装无载体纳米颗粒(GEM-CXBNP)开发了一种新方法,用于乳腺癌化学免疫疗法中MDSC和肿瘤细胞的双重消耗。GEM-CXBNP表现出延长的血液循环,导致GEM和CXB在肿瘤中优先积累和共同释放。这通过对4T1肿瘤细胞的增殖抑制和凋亡诱导来促进协同化疗活性。此外,它通过免疫原性细胞死亡诱导和MDSC诱导的免疫抑制通过消耗MDSC增强肿瘤免疫原性。这些机制协同激活细胞毒性T细胞和自然杀伤细胞的抗肿瘤免疫功能,抑制调节性T细胞的增殖,并促进肿瘤相关巨噬细胞的M2至M1表型复极化,显着增强了携带4T1肿瘤的BALB/c小鼠的整体抗肿瘤和抗转移功效。GEM-CXBNP的简化工程,以免疫抑制细胞和肿瘤细胞为目标的双重消耗策略,代表了癌症化学免疫疗法的先进概念。
    Myeloid-derived suppressor cells (MDSCs) have played a significant role in facilitating tumor immune escape and inducing an immunosuppressive tumor microenvironment. Eliminating MDSCs and tumor cells remains a major challenge in cancer immunotherapy. A novel approach has been developed using gemcitabine-celecoxib twin drug-based nano-assembled carrier-free nanoparticles (GEM-CXB NPs) for dual depletion of MDSCs and tumor cells in breast cancer chemoimmunotherapy. The GEM-CXB NPs exhibit prolonged blood circulation, leading to the preferential accumulation and co-release of GEM and CXB in tumors. This promotes synergistic chemotherapeutic activity by the proliferation inhibition and apoptosis induction against 4T1 tumor cells. In addition, it enhances tumor immunogenicity by immunogenic cell death induction and MDSC-induced immunosuppression alleviation through the depletion of MDSCs. These mechanisms synergistically activate the antitumor immune function of cytotoxic T cells and natural killer cells, inhibit the proliferation of regulatory T cells, and promote the M2 to M1 phenotype repolarization of tumor-associated macrophages, considerably enhancing the overall antitumor and anti-metastasis efficacy in BALB/c mice bearing 4T1 tumors. The simplified engineering of GEM-CXB NPs, with their dual depletion strategy targeting immunosuppressive cells and tumor cells, represents an advanced concept in cancer chemoimmunotherapy.
    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
    转移性胰腺癌(mPC)预后不良。在这里,我们进行了一个前瞻性的,多中心,单臂,II期试验评估pepulimab和安洛替尼联合nab-紫杉醇/吉西他滨(PAAG)在一线mPC患者中的疗效和安全性(NCT05493995).主要终点包括客观缓解率(ORR)和疾病控制率(DCR)。而次要终点包括无进展生存期(PFS),总生存期(OS),和安全。在分析疗效的66例患者中,最好的回应,由ORR指示,记录为50.0%(33/66)(95%CI,37.4-62.6%),33例患者达到部分缓解(PR)。值得注意的是,DCR为95.5%(63/66,95%CI,87.3-99.1%)。中位PFS(mPFS)和OS(mOS)为8.8(95%CI,8.1-11.6),和13.7个月(95%CI,12.4至未达到),分别。39.4%的患者报告了3/4级治疗相关不良事件(TRAEs)(26/66)。在预先指定的探索性分析中,SWI/SNF复合物改变的患者PFS较差.此外,低血清CA724水平,高T细胞募集,低Th17细胞募集,基线时较高的NKCD56dim细胞评分是疗效更有利的潜在预测生物标志物.总之,PAAG作为一线治疗证明了mPC的耐受性和有希望的临床疗效。本研究中确定的生物分子发现具有指导三重组合方案的精确临床应用的潜力。
    Metastatic pancreatic cancer (mPC) has a dismal prognosis. Herein, we conducted a prospective, multicentre, single-arm, phase II trial evaluating the efficacy and safety of penpulimab and anlotinib in combination with nab-paclitaxel/gemcitabine (PAAG) in patients with first-line mPC (NCT05493995). The primary endpoints included the objective response rate (ORR) and disease control rate (DCR), while secondary endpoints encompassed progression-free survival (PFS), overall survival (OS), and safety. In 66 patients analysed for efficacy, the best response, indicated by the ORR, was recorded at 50.0% (33/66) (95% CI, 37.4-62.6%), with 33 patients achieving partial response (PR). Notably, the DCR was 95.5% (63/66, 95% CI, 87.3-99.1%). The median PFS (mPFS) and OS (mOS) were 8.8 (95% CI, 8.1-11.6), and 13.7 (95% CI, 12.4 to not reached) months, respectively. Grade 3/4 treatment-related adverse events (TRAEs) were reported in 39.4% of patients (26/66). In prespecified exploratory analysis, patients with altered SWI/SNF complex had a poorer PFS. Additionally, low serum CA724 level, high T-cell recruitment, low Th17 cell recruitment, and high NK CD56dim cell scores at baseline were potential predicative biomarkers for more favourable efficacy. In conclusion, PAAG as a first-line therapy demonstrated tolerability with promising clinical efficacy for mPC. The biomolecular findings identified in this study possess the potential to guide the precise clinical application of the triple-combo regimen.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号