Deoxycytidine

脱氧胞苷
  • 文章类型: Journal Article
    胰腺导管腺癌(PDAC)是胰腺癌的最常见形式,一种总体生存率很低的疾病。缺乏临床前模型阻碍了治疗的进展。这里,我们展示了从切除组织创建的个性化器官型\"化身\"如何允许对完整的原位肿瘤微环境进行空间和时间报告,并反映临床反应.我们的灌注培养方法扩展了肿瘤切片的活力,保持稳定的肿瘤含量,新陈代谢,基质成分,和免疫细胞群持续12天。使用多重免疫荧光和空间转录组学,我们确定了免疫邻域和免疫疗法的潜力。我们使用化身来评估临床前验证的代谢疗法的影响,并显示基质和免疫表型的恢复以及肿瘤再分化。为了确定临床相关性,我们监测了患者对吉西他滨治疗的化身反应,并从临床随访中确定了患者化身可预测的反应.因此,化身为治疗方法的同基因测试提供了有价值的信息,并为患者提供了一个真正个性化的治疗评估平台。
    Pancreatic ductal adenocarcinoma (PDAC) is the most common form of pancreatic cancer, a disease with dismal overall survival. Advances in treatment are hindered by a lack of preclinical models. Here, we show how a personalized organotypic \"avatar\" created from resected tissue allows spatial and temporal reporting on a complete in situ tumor microenvironment and mirrors clinical responses. Our perfusion culture method extends tumor slice viability, maintaining stable tumor content, metabolism, stromal composition, and immune cell populations for 12 days. Using multiplexed immunofluorescence and spatial transcriptomics, we identify immune neighborhoods and potential for immunotherapy. We used avatars to assess the impact of a preclinically validated metabolic therapy and show recovery of stromal and immune phenotypes and tumor redifferentiation. To determine clinical relevance, we monitored avatar response to gemcitabine treatment and identify a patient avatar-predictable response from clinical follow-up. Thus, avatars provide valuable information for syngeneic testing of therapeutics and a truly personalized therapeutic assessment platform for patients.
    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
    吉西他滨加nab-紫杉醇(GnP)和FOLFIRINOX被广泛用作不可切除的胰腺癌(PC)的一线方案。当选择GnP治疗时,考虑到患者的年龄或状况,二线FOLFIRINOX由于其毒性有时难以给药。本研究旨在确定S-IROX(S-1,奥沙利铂,和伊立替康联合用药)一线GnP失败后无法切除的PC患者的治疗方案。该I期研究使用了两种剂量水平的“3+3”剂量递增设计。纳入一线GnP治疗无法切除的PC失败的患者。在第1天施用奥沙利铂和伊立替康,并且在第1-7天每天两次口服施用S-1,随后休息7天。主要终点是剂量限制性毒性(DLTs)和RD的测定。次要终点是潜在抗肿瘤活性的评估。9例患者接受了二线S-IROX方案。在0级(S-1,80毫克/平方米;奥沙利铂,85毫克/平方米;伊立替康,120mg/m2),没有患者经历DLT;然而,1例患者出现3级中性粒细胞减少症.在1级(伊立替康增加到150毫克/平方米),六名患者中有一名经历了DLT,包括G3腹泻。RD在1级剂量下得到证实。响应率,疾病控制率,中位无进展生存期,中位总生存率为33.3%,77.8%,172(范围:77-422)天,和414(101-685)天,分别。一名患者在二线S-IROX治疗后接受了手术。二线S-IROX治疗被认为是可接受的。RD设定为1级剂量(S-1,80mg/m2;奥沙利铂,85毫克/平方米;伊立替康,150mg/m2)。
    Gemcitabine plus nab-paclitaxel (GnP) and FOLFIRINOX are widely used as first-line regimens for unresectable pancreatic cancer (PC). When GnP therapy is selected, considering patient age or condition, second-line FOLFIRINOX is sometimes difficult to administer owing to its toxicity. This study aimed to determine the recommended dose (RD) of S-IROX (S-1, oxaliplatin, and irinotecan combination) regimens in patients with unresectable PC after first-line GnP failure. This phase-I study used the \"3 + 3\" dose-escalation design with two dose levels. Patients who failed first-line GnP therapy for unresectable PC were enrolled. Oxaliplatin and irinotecan were administered on day 1, and S-1 was administered orally twice daily on days 1-7, followed by 7 days of rest. The primary endpoints were dose-limiting toxicities (DLTs) and determination of RD. The secondary endpoint was the evaluation of potential antitumor activity. Nine patients received the second-line S-IROX regimen. In level-0 (S-1, 80 mg/m2; oxaliplatin, 85 mg/m2; and irinotecan, 120 mg/m2), no patient experienced DLT; however, one patient experienced grade 3 neutropenia. At level-1 (irinotecan increased to 150 mg/m2), one of six patients experienced DLTs, including G3 diarrhea. The RD was confirmed at the level-1 dose. The response rate, disease control rate, median progression-free survival, and median overall survival were 33.3%, 77.8%, 172 (range:77-422) days, and 414 (101-685) days, respectively. One patient underwent surgery after the second-line S-IROX therapy. Second-line S-IROX treatment was deemed acceptable. The RD was set at level-1 dose (S-1, 80 mg/m2; oxaliplatin, 85 mg/m2; and irinotecan, 150 mg/m2).
    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

    求助全文

  • 文章类型: Journal Article
    胰腺导管腺癌(PDAC)的特征是纤维化增加,可以促进肿瘤的进展和扩散。这里,我们对高转移性KPC(Pdx1-Cre,LSL-KrasG12D/+,LSL-Trp53R172H/+)和低转移性KPflC(Pdx1-Cre,LSL-KrasG12D/+,Trp53fl/+)使用质谱蛋白质组学对胰腺癌进行基因工程小鼠模型。我们在早期的评估-,mid-,和晚期疾病揭示了与KPflC相比,KPC模型中nidogen-2(NID2)的丰度增加,进一步验证显示NID2主要由癌症相关成纤维细胞(CAF)表达。使用生物力学评估,二次谐波产生成像,和双折射分析,我们表明,在CAF中通过CRISPR干扰(CRISPRi)减少NID2降低了三维模型中的刚度和基质重塑,导致癌细胞侵袭受损。活体成像显示NID2耗尽的活体肿瘤血管通畅改善,对吉西他滨/Abraxane的反应增强。在原位模型中,NID2CRISPRi肿瘤肝转移较少,生存率增加,突出显示NID2作为潜在的PDAC共目标。
    Pancreatic ductal adenocarcinoma (PDAC) is characterized by increasing fibrosis, which can enhance tumor progression and spread. Here, we undertook an unbiased temporal assessment of the matrisome of the highly metastatic KPC (Pdx1-Cre, LSL-KrasG12D/+, LSL-Trp53R172H/+) and poorly metastatic KPflC (Pdx1-Cre, LSL-KrasG12D/+, Trp53fl/+) genetically engineered mouse models of pancreatic cancer using mass spectrometry proteomics. Our assessment at early-, mid-, and late-stage disease reveals an increased abundance of nidogen-2 (NID2) in the KPC model compared to KPflC, with further validation showing that NID2 is primarily expressed by cancer-associated fibroblasts (CAFs). Using biomechanical assessments, second harmonic generation imaging, and birefringence analysis, we show that NID2 reduction by CRISPR interference (CRISPRi) in CAFs reduces stiffness and matrix remodeling in three-dimensional models, leading to impaired cancer cell invasion. Intravital imaging revealed improved vascular patency in live NID2-depleted tumors, with enhanced response to gemcitabine/Abraxane. In orthotopic models, NID2 CRISPRi tumors had less liver metastasis and increased survival, highlighting NID2 as a potential PDAC cotarget.
    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
    背景和目的:吉西他滨已被用于治疗各种实体癌,包括,自1997年以来,转移性胰腺癌。这里,我们开发了HPLC-UV法测定血清吉西他滨水平,并将其用于药代动力学研究.材料和方法:分析是在反相柱上进行单个蛋白沉淀步骤后进行的,用磷酸钠缓冲液和甲醇等度洗脱。对于药代动力学研究,NOD/SCID小鼠通过皮下(SC)或腹膜内(IP)施用以100mg/kg接受单剂量的吉西他滨。在施用吉西他滨后5、15和30分钟以及1、2、4和6小时收集血样用于进一步分析。结果:分析的持续时间为~12.5分钟。校准曲线在1-400μM的范围内是线性的(r2=0.999)。GEM的平均回收率为96.53%,检测限为0.166μΜ。T1/2,Tmax,Cmax,AUC0-t,清除率为64.49分钟,5.00分钟,264.88μmol/L,9351.95μmol/L*min,0.0103(mg)/(μmol/L)/min,分别,SC管理。IP管理的相应值为59.34分钟,5.00分钟,300.73μmol/L,8981.35μmol/L*min和0.0108(mg)/(μmol/L)/min(与SC给药没有统计学差异)。结论:一个简单的,有效,敏感,已经开发了用于测量血清中吉西他滨的廉价方法。该方法可用于监测癌症患者的吉西他滨水平,作为治疗药物监测的一部分。
    Background and Objectives: Gemcitabine has been used to treat various solid cancers, including, since 1997, metastatic pancreatic cancer. Here, we developed an HPLC-UV method to determine serum gemcitabine levels and use it in pharmacokinetic studies. Materials and Methods: The analysis was performed after a single protein precipitation step on a reversed-phase column, isocratically eluted with sodium phosphate buffer and methanol. For the pharmacokinetic study, NOD/SCID mice received a single dose of gemcitabine at 100 mg/kg by either subcutaneous (SC) or intraperitoneal (IP) administration. Blood samples were collected at 5, 15, and 30 min and 1, 2, 4, and 6 h after the administration of gemcitabine for further analysis. Results: The duration of the analysis was ~12.5 min. The calibration curve was linear (r2 = 0.999) over the range of 1-400 μM. The mean recovery of GEM was 96.53% and the limit of detection was 0.166 μΜ. T1/2, Tmax, Cmax, AUC0-t, and clearance were 64.49 min, 5.00 min, 264.88 μmol/L, 9351.95 μmol/L*min, and 0.0103(mg)/(μmol/L)/min, respectively, for the SC administration. The corresponding values for the IP administration were 59.34 min, 5.00 min, 300.73 μmol/L, 8981.35 μmol/L*min and 0.0108(mg)/(μmol/L)/min (not statistically different from the SC administration). Conclusions: A simple, valid, sensitive, and inexpensive method for the measurement of gemcitabine in serum has been developed. This method may be useful for monitoring gemcitabine levels in cancer patients as part of therapeutic drug monitoring.
    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:基于吉西他滨(GEM)的化疗已被确立为胆道癌(BTC)的核心多模式治疗。然而,由于BTC对GEM的抗性,其预后不利。外泌体在调节肿瘤的进展和转移中起重要作用,免疫失调,和化学抗性。本研究调查了外泌体对BTCGEM抗性的影响。
    方法:人肝内胆管癌细胞系CC-LP-1,其GEM抗性(GR)衍生细胞系CC-LP-1-GR,使用人肝内胆管癌细胞系HuCCA-1和HuCCT1。通过使用MTS测定在GEM存在下测量细胞活力来检查GEM抗性。使用超速离心分离外泌体并使用ELISA定量。使用RNA测序进行综合表达分析。通过miRNA模拟物转染检查微小RNA的作用。
    结果:条件培养基和源自CC-LP-1-GR细胞的外泌体增强了亲本CC-LP-1细胞的GEM抗性。在创业板的存在下,与CC-LP-1细胞相比,用CC-LP-1-GR(rExo)外泌体处理的CC-LP-1-GR和CC-LP-1细胞中p53通路呈负富集.rExos中miR-141-3p的表达高于CC-LP-1细胞。用miR-141-3p模拟物转染的CC-LP-1细胞显示在GEM存在下显著(p<0.05)增加的活力。
    结论:GEM抗性人BTC细胞系,CC-LP-1-GR,可能通过含有miR-141-3p的外泌体获得对GEM的抗性。
    OBJECTIVE: Gemcitabine (GEM)-based chemotherapy has been established as the core multimodal therapy for biliary tract cancer (BTC). However, the prognosis of BTC is unfavorable because of its resistance to GEM. Exosomes play important roles in the regulation of tumor progression and metastasis, immune dysregulation, and chemoresistance. This study investigated the effects of exosomes on GEM resistance in BTC.
    METHODS: The human intrahepatic cholangiocarcinoma cell line CC-LP-1, its GEM-resistant (GR) derivative cell line CC-LP-1-GR, and the human intrahepatic cholangiocarcinoma cell lines HuCCA-1 and HuCCT1, were used. GEM resistance was examined by measuring cell viability in the presence of GEM using an MTS assay. Exosomes were isolated using ultracentrifugation and quantified using ELISA. Comprehensive expression analysis was performed using RNA sequencing. The effects of microRNAs were examined by miRNA mimic transfection.
    RESULTS: The conditioned medium and exosomes derived from CC-LP-1-GR cells enhanced the GEM resistance of parental CC-LP-1 cells. In the presence of GEM, the p53 pathway was negatively enriched in CC-LP-1-GR and CC-LP-1 cells treated with exosomes from CC-LP-1-GR (rExo) compared to CC-LP-1 cells. The expression of miR-141-3p was higher in rExos than in CC-LP-1 cells. CC-LP-1 cells transfected with miR-141-3p mimic showed significantly (p<0.05) increased viability in the presence of GEM.
    CONCLUSIONS: A GEM-resistant human BTC cell line, CC-LP-1-GR, may acquire resistance to GEM by exosomes containing miR-141-3p.
    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:胰腺癌(PC)一线治疗通常包括多化疗方案,但是在疾病进展后选择二线治疗,尤其是在一线FOLFIRINOX之后,仍然是一个临床挑战。这项研究提出了一个大的结果,多中心,回顾性分析接受Nab-紫杉醇/吉西他滨(AG)作为二线或后期治疗的意大利转移性PC(mPC)患者.该研究的主要目的是确定可以为治疗决策提供信息的预后因素。
    方法:该研究包括在17个意大利机构接受AG治疗的160名mPC患者。AG根据标签剂量给药,直到疾病进展,不可接受的毒性或患者拒绝。时间表的变化,剂量修改,支持性措施,和反应评估由个别临床医生实践确定。
    结果:AG具有良好的耐受性并表现出良好的临床活性。总有效率(ORR)和疾病控制率(DCR)分别为22.5%和45.6%,分别。中位无进展生存期(PFS)和总生存期(OS)分别为3.9和6.8个月,分别。在接受AG作为二线治疗的患者中(n=111,66.9%),中位PFS和OS分别为4.2和7.4个月,分别。值得注意的是,在一线FOLFIRINOX后接受AG的76例患者(68%)中,观察到19.7%的ORR和46.0%的DCR,导致中位PFS为3.5个月,中位OS为5.7个月。该研究确定了特定的临床或实验室参数(LDH,NLR,空腹血糖,肝转移,ECOGPS,和一线PFS)作为多变量水平的独立预后因素。这些因素被用来创建一个预后列线图,将患者分为三个风险等级,帮助预测二线OS和PFS。
    结论:这项研究代表了接受AG作为第二或更晚治疗的mPC患者的最大现实世界人群。它支持该方案在一线FOLFIRINOX之后的可行性,特别是在具有特定临床和实验室特征并从一线治疗中获得长期益处的患者中。
    BACKGROUND: Pancreatic cancer (PC) first-line therapy often consists of polychemotherapy regimens, but choosing a second-line therapy after disease progression, especially following first-line FOLFIRINOX, remains a clinical challenge. This study presents results from a large, multicenter, retrospective analysis of Italian patients with metastatic PC (mPC) treated with Nab-paclitaxel/Gemcitabine (AG) as second or later line of treatment. Main objective of the study is to identify prognostic factors that could inform treatment decisions.
    METHODS: The study included 160 mPC patients treated with AG in 17 Italian institutions. AG was administered according to labelling dose, until disease progression, unacceptable toxicity or patient refusal. Variations in schedules, dose modifications, supportive measures, and response evaluation were determined by individual clinicians\' practice.
    RESULTS: AG was well-tolerated and exhibited promising clinical activity. The overall response rate (ORR) and the disease control rate (DCR) were 22.5% and 45.6%, respectively. Median progression-free survival (PFS) and overall survival (OS) were 3.9 and 6.8 months, respectively. Among the patients who received AG as a second-line therapy (n = 111, 66.9%), median PFS and OS were 4.2 and 7.4 months, respectively. Notably, in the 76 patients (68%) receiving AG after first-line FOLFIRINOX, an ORR of 19.7% and a DCR of 46.0% were observed, resulting in a median PFS of 3.5 and median OS of 5.7 months. The study identified specific clinical or laboratory parameters (LDH, NLR, fasting serum glucose, liver metastases, ECOG PS, and first-line PFS) as independent prognostic factors at multivariate level. These factors were used to create a prognostic nomogram that divided patients into three risk classes, helping to predict second-line OS and PFS.
    CONCLUSIONS: This study represents the largest real-world population of mPC patients treated with AG as a second or later line of therapy. It supports the feasibility of this regimen following first-line FOLFIRINOX, particularly in patients with specific clinical and laboratory characteristics who derived prolonged benefit from first-line 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
    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
    胆管癌(CCA)是起源于胆管上皮的侵袭性癌症,在亚洲国家尤其普遍的肝吸虫感染。目前用于CCA的化疗通常由于耐药性而失败,需要新的抗癌剂。本研究调查了5'-脱氧-5'-甲硫腺苷(MTA)的潜力,一种天然存在的核苷,反对CCA。虽然MTA已经显示出对抗各种癌症的希望,其对CCA的影响仍未被探索。我们评估了MTA在CCA细胞系和耐药亚系中的抗癌活性,评估细胞活力,迁移,入侵,和凋亡。通过使用LC-MS/MS的蛋白质组学分析和生物信息学分析,探索了MTA的潜在抗癌机制。结果显示CCA细胞活力的剂量依赖性降低,与正常细胞相比,对癌细胞的作用增强。此外,MTA抑制生长,诱导细胞凋亡,并抑制细胞迁移和侵袭。此外,MTA增强吉西他滨对耐药CCA细胞的抗癌作用。蛋白质组学揭示了MTA对多种蛋白质的下调,影响各种分子功能,生物过程,和细胞成分。网络分析强调MTA在抑制与线粒体功能和能量衍生相关的蛋白质中的作用,对细胞生长和存活至关重要。此外,MTA抑制参与细胞形态和细胞骨架组织的蛋白质,重要的癌细胞运动和转移。六个候选基因,包括ZNF860,KLC1,GRAMD1C,妈妈,TANC1和TTC13选自蛋白质组学结果中鉴定的前10种最下调的蛋白质,随后通过RT-qPCR验证。Further,通过蛋白质印迹证实MTA处理对KLC1蛋白的抑制。此外,基于TCGA数据,与正常邻近组织相比,CCA患者的组织中KLC1mRNA被上调。总之,MTA通过抑制生长显示出对CCA的有希望的抗癌潜力,诱导细胞凋亡,抑制迁移和入侵,同时增强吉西他滨的效果。蛋白质组学分析阐明了MTA抗癌活性潜在的分子机制,为未来MTA作为先进CCA治疗方法的研究和发展奠定基础。
    Cholangiocarcinoma (CCA) is an aggressive cancer originating from bile duct epithelium, particularly prevalent in Asian countries with liver fluke infections. Current chemotherapy for CCA often fails due to drug resistance, necessitating novel anticancer agents. This study investigates the potential of 5\'-deoxy-5\'-methylthioadenosine (MTA), a naturally occurring nucleoside, against CCA. While MTA has shown promise against various cancers, its effects on CCA remain unexplored. We evaluated MTA\'s anticancer activity in CCA cell lines and drug-resistant sub-lines, assessing cell viability, migration, invasion, and apoptosis. The potential anticancer mechanisms of MTA were explored through proteomic analysis using LC-MS/MS and bioinformatic analysis. The results show a dose-dependent reduction in CCA cell viability, with enhanced effects on cancer cells compared to normal cells. Moreover, MTA inhibits growth, induces apoptosis, and suppresses cell migration and invasion. Additionally, MTA enhanced the anticancer effects of gemcitabine on drug-resistant CCA cells. Proteomics revealed the down-regulation of multiple proteins by MTA, affecting various molecular functions, biological processes, and cellular components. Network analysis highlighted MTA\'s role in inhibiting proteins related to mitochondrial function and energy derivation, crucial for cell growth and survival. Additionally, MTA suppressed proteins involved in cell morphology and cytoskeleton organization, important for cancer cell motility and metastasis. Six candidate genes, including ZNF860, KLC1, GRAMD1C, MAMSTR, TANC1, and TTC13, were selected from the top 10 most down-regulated proteins identified in the proteomics results and were subsequently verified through RT-qPCR. Further, KLC1 protein suppression by MTA treatment was confirmed through Western blotting. Additionally, based on TCGA data, KLC1 mRNA was found to be upregulated in the tissue of CCA patients compared to that of normal adjacent tissues. In summary, MTA shows promising anticancer potential against CCA by inhibiting growth, inducing apoptosis, and suppressing migration and invasion, while enhancing gemcitabine\'s effects. Proteomic analysis elucidates possible molecular mechanisms underlying MTA\'s anticancer activity, laying the groundwork for future research and development of MTA as a treatment for advanced CCA.
    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)

公众号