Pancreatic neoplasms

胰腺肿瘤
  • 文章类型: Journal Article
    胰腺导管腺癌是全球预后最差的实体瘤之一,辅助或新辅助治疗后复发率高。循环肿瘤DNA分析是表征肿瘤基因组学和评估治疗反应的一种有前途的非侵入性工具。在这项研究中,手术肿瘤组织和序贯血液样本通过下一代测序进行分析,并与临床和病理特征相关.包括在纳瓦拉大学医院接受治疗的30名可切除/交界性胰腺导管腺癌患者。循环肿瘤DNA测序鉴定了KRAS和TP53以及其他癌症相关基因的致病变异。诊断时在预后较差的患者中检测到致病性变异,并且与临界胰腺导管型肾上腺癌患者对新辅助治疗的反应相关。诊断时较高的变异等位基因频率与较差的预后相关,在进展时,样本中变异等位基因频率的总和更大。我们的结果建立在非转移性胰腺导管腺癌患者循环肿瘤DNA的潜在价值,通过补充组织遗传信息,并作为治疗决策的非侵入性工具。需要进行验证性研究来证实这些发现。
    Pancreatic ductal adenocarcinoma represents one of the solid tumors showing the worst prognosis worldwide, with a high recurrence rate after adjuvant or neoadjuvant therapy. Circulating tumor DNA analysis raised as a promising non-invasive tool to characterize tumor genomics and to assess treatment response. In this study, surgical tumor tissue and sequential blood samples were analyzed by next-generation sequencing and were correlated with clinical and pathological characteristics. Thirty resectable/borderline pancreatic ductal adenocarcinoma patients treated at the Hospital Universitario de Navarra were included. Circulating tumoral DNA sequencing identified pathogenic variants in KRAS and TP53, and in other cancer-associated genes. Pathogenic variants at diagnosis were detected in patients with a poorer outcome, and were correlated with response to neoadjuvant therapy in borderline pancreatic ductal adneocarcinoma patients. Higher variant allele frequency at diagnosis was associated with worse prognosis, and thesum of variant allele frequency was greater in samples at progression. Our results build on the potential value of circulating tumor DNA for non-metastatic pancreatic ductal adenocarcinoma patients, by complementing tissue genetic information and as a non-invasive tool for treatment decision. Confirmatory studies are needed to corroborate these findings.
    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
    越来越多的证据表明,生物系统在结构和功能上都是模块化的。复杂的生物信号网络,如基因调控网络(GRN)被证明是由相互联系和分层排列的子类别组成的。这些网络包含高度动态的过程,这些过程最终决定了细胞功能。以及影响表型命运转变。在这项工作中,我们使用胰腺癌(PC)的随机多细胞信号网络显示,最具表型影响的模块的拓扑排序的差异意味着结构和功能之间有很强的关系.我们进一步表明,突变的诱导改变了模块化结构,这类似地影响计算机中疾病的攻击性和可控性。最后,我们提出了证据,表明突变对PC模块化结构的影响和位置直接对应于硅片中单剂治疗的功效,因为拓扑深层突变需要深层目标来控制。
    There is increasing evidence that biological systems are modular in both structure and function. Complex biological signaling networks such as gene regulatory networks (GRNs) are proving to be composed of subcategories that are interconnected and hierarchically ranked. These networks contain highly dynamic processes that ultimately dictate cellular function over time, as well as influence phenotypic fate transitions. In this work, we use a stochastic multicellular signaling network of pancreatic cancer (PC) to show that the variance in topological rankings of the most phenotypically influential modules implies a strong relationship between structure and function. We further show that induction of mutations alters the modular structure, which analogously influences the aggression and controllability of the disease in silico. We finally present evidence that the impact and location of mutations with respect to PC modular structure directly corresponds to the efficacy of single agent treatments in silico, because topologically deep mutations require deep targets for control.
    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
    目的:罕见胰腺肿瘤的生物学,不同于导管胰腺癌,需要更多的关注。尽管大多数罕见的胰腺肿瘤是良性的,如果不完全切除病变,很难确定是否存在侵入性成分,尽管在诊断方面取得了相当大的进展。我们正在调查大量经组织学证实的胰腺上皮非导管非神经内分泌肿瘤患者。
    方法:在这里,我们分析了患者的长期生存率,接受组织学证实的胰腺上皮非导管非神经内分泌肿瘤切除术的患者。1月1日之间在我们部门,1999年和12月31日,2019.中位随访时间为61(范围0-168)个月。所有统计分析均使用SPSS26.0(IBM,芝加哥,IL,美国)软件。
    结果:46例(48%)随访5年以上,18例(19%)患者10年以上。罕见的非侵袭性胰腺肿瘤的5年和10年生存率分别为72%和55%。在1999年至2019年期间,我们的诊所中罕见肿瘤实体(非导管和非神经内分泌)的比例从4.2%连续增加到12.3%(p=0.004)。如果还没有侵入性生长,在疾病的过程中,恶性变性的风险各不相同。因此,胰腺切除的适应症仍是讨论的主题.
    结论:R0切除后的罕见上皮性胰腺肿瘤的长期预后-即使它们已经是恶性的-比导管胰腺癌的预后要好得多。
    OBJECTIVE: The biology of rare pancreatic tumours, which differs from that of ductal pancreatic cancer, requires increased attention. Although the majority of rare pancreatic tumours are benign, it is difficult to decide whether an invasive component exists without complete removal of the lesion, despite considerable progress in diagnosis. We are investigating a large cohort of patients with histologically confirmed epithelial non-ductal non-neuroendocrine neoplasms of the pancreas.
    METHODS: Here we analyze long-term survival from patients, who underwent resection of histologically confirmed epithelial non-ductal non-neuroendocrine neoplasms of the pancreas. At our department between Jan 1st, 1999, and Dec 31st, 2019. The median follow-up was 61 (range 0-168) month. All statistical analyses were performed using SPSS 26.0 (IBM, Chicago, IL, USA) software.
    RESULTS: 46 patients (48%) were followed up for more than 5 years, 18 patients (19%) for more than 10 years. The 5-year and 10-year survival rates for rare non-invasive pancreatic tumours were 72% and 55% respectively. The proportion of rare tumour entities (non-ductal and non-neuroendocrine) increased continuously and statistically significantly (p = 0.004) from 4.2 to 12.3% in our clinic between 1999 and 2019. If there is no invasive growth yet, there is a varying risk of malignant degeneration in the course of the disease. Therefore, the indication for pancreatic resection is still the subject of discussion.
    CONCLUSIONS: The long-term prognosis of rare epithelial pancreatic tumours after R0 resection-even if they are already malignant-is much better than that of ductal 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
    胰腺癌仍然是一个重要的健康问题,治疗选择有限。肿瘤间质,由不同的细胞和蛋白质组成的复杂环境,在肿瘤生长和化疗耐药中起着至关重要的作用。靶向肿瘤基质,由不同的非肿瘤细胞如成纤维细胞组成,细胞外基质(ECM),免疫细胞,血管前细胞也鼓励重塑实体癌,比如胰腺癌。重塑胰腺肿瘤的基质可以被建议作为降低对化学/免疫疗法的抗性的策略。一些研究表明,来自植物的植物化学物质可以影响肿瘤环境并具有抗癌特性。通过靶向参与基质激活的关键途径,植物化学物质可能会破坏肿瘤和间质之间的交流,使肿瘤细胞对不同的治疗方法更敏感。此外,植物化学物质具有免疫调节和抗血管生成特性,所有这些都有助于他们治疗胰腺癌的潜力。这篇综述将详细介绍植物化学物质如何影响肿瘤基质及其对胰腺肿瘤生长的影响。传播,以及对治疗的反应。它还将探索将植物化学物质与化疗等其他治疗方案相结合的潜力,免疫疗法,和辐射。
    Pancreatic cancer remains a significant health issue with limited treatment options. The tumor stroma, a complex environment made up of different cells and proteins, plays a crucial role in tumor growth and chemoresistance. Targeting tumor stroma, consisting of diverse non-tumor cells such as fibroblasts, extracellular matrix (ECM), immune cells, and also pre-vascular cells is encouraging for remodeling solid cancers, such as pancreatic cancer. Remodeling the stroma of pancreas tumors can be suggested as a strategy for reducing resistance to chemo/immunotherapy. Several studies have shown that phytochemicals from plants can affect the tumor environment and have anti-cancer properties. By targeting key pathways involved in stromal activation, phytochemicals may disrupt communication between the tumor and stroma and make tumor cells more sensitive to different treatments. Additionally, phytochemicals have immunomodulatory and anti-angiogenic properties, all of which contribute to their potential in treating pancreatic cancer. This review will provide a detailed look at how phytochemicals impact the tumor stroma and their effects on pancreatic tumor growth, spread, and response to treatment. It will also explore the potential of combining phytochemicals with other treatment options like chemotherapy, immunotherapy, and radiation.
    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
    针对胰腺导管腺癌(PDAC)的化疗治疗因多种治疗抵抗途径的肿瘤激活而受阻。生长激素(GH)-GH受体(GHR)对是癌症多模式治疗抵抗的隐性驱动因素,并且在PDAC肿瘤中过度表达。然而,靶向相同的治疗潜力尚未被探索。这里,我们报道GHR的表达是PDAC患者的不良预后因素.吉西他滨与不同GHR拮抗剂(GHRA)的组合显著改善裸鼠异种移植物的治疗结果。使用培养的细胞,小鼠异种移植物,以及人类PDAC转录组的分析,我们发现,肿瘤中多药转运体和上皮-间质转化程序的减弱是GHRAs观察到的化疗疗效增强的基础.此外,在人类PDAC患者中,GHR表达与肿瘤促进和免疫逃避的基因签名密切相关,这证实了野生型与同基因肿瘤的关系。GH转基因小鼠。总的来说,我们发现GH在PDAC中的作用促进了体内治疗难治性基因签名,可以通过GHR拮抗作用有效减弱。我们的结果共同证明了在高度化学抗性PDAC中考虑GHR拮抗剂以改善化疗结果的概念。
    Chemotherapy treatment against pancreatic ductal adenocarcinoma (PDAC) is thwarted by tumoral activation of multiple therapy resistance pathways. The growth hormone (GH)-GH receptor (GHR) pair is a covert driver of multimodal therapy resistance in cancer and is overexpressed in PDAC tumors, yet the therapeutic potential of targeting the same has not been explored. Here, we report that GHR expression is a negative prognostic factor in patients with PDAC. Combinations of gemcitabine with different GHR antagonists (GHRAs) markedly improve therapeutic outcomes in nude mice xenografts. Employing cultured cells, mouse xenografts, and analyses of the human PDAC transcriptome, we identified that attenuation of the multidrug transporter and epithelial-to-mesenchymal transition programs in the tumors underlie the observed augmentation of chemotherapy efficacy by GHRAs. Moreover, in human PDAC patients, GHR expression strongly correlates with a gene signature of tumor promotion and immune evasion, which corroborate with that in syngeneic tumors in wild-type vs. GH transgenic mice. Overall, we found that GH action in PDAC promoted a therapy-refractory gene signature in vivo, which can be effectively attenuated by GHR antagonism. Our results collectively present a proof of concept toward considering GHR antagonists to improve chemotherapeutic outcomes in the highly chemoresistant 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
    表观遗传变化在癌症中很常见,包括异常的DNA甲基化和组蛋白修饰。包括乙酰化或甲基化。启动子区域的DNA甲基化和组蛋白脱乙酰通常伴随着基因沉默,并可能导致癌细胞中肿瘤抑制因子的抑制。据报道,表观遗传途径之间的相互作用可用于更有效地靶向侵袭性癌细胞。特别是那些目前的治疗通常失败,比如胰腺癌。在这项研究中,我们探索了将DNA去甲基化剂5-AZA与HDAC抑制剂SAHA联合治疗胰腺癌细胞系的可能性,专注于mutp53的乙酰化及其稳定性的后果,以及该蛋白与c-myc和BRCA-1的相互作用,这些是癌症生存的关键分子。获得的结果表明,SAHA/5-AZA组合比单一治疗更有效地促进mutp53的降解,上调p21并下调c-Myc和BRCA-1,从而增加胰腺癌细胞的DNA损伤和细胞毒性。
    Epigenetic changes are common in cancer and include aberrant DNA methylation and histone modifications, including both acetylation or methylation. DNA methylation in the promoter regions and histone deacetylation are usually accompanied by gene silencing, and may lead to the suppression of tumor suppressors in cancer cells. An interaction between epigenetic pathways has been reported that could be exploited to more efficiently target aggressive cancer cells, particularly those against which current treatments usually fail, such as pancreatic cancer. In this study, we explored the possibility to combine the DNA demethylating agent 5-AZA with HDAC inhibitor SAHA to treat pancreatic cancer cell lines, focusing on the acetylation of mutp53 and the consequences on its stability, as well as on the interaction of this protein with c-myc and BRCA-1, key molecules in cancer survival. The results obtained suggest that SAHA/5-AZA combination was more effective than single treatments to promote the degradation of mutp53, to upregulate p21 and downregulate c-Myc and BRCA-1, thus increasing DNA damage and cytotoxicity in pancreatic cancer cells.
    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)在诊断和治疗方面提出了重大挑战,治疗选择有限,预后不良。这项研究探索了NPS-1034的潜在治疗作用,一种针对MET和AXL的激酶抑制剂,在PDAC。研究包括NPS-1034的单药治疗及其与常用化疗药物的联合治疗。氟尿嘧啶和奥沙利铂。我们的研究表明,NPS-1034以剂量依赖的方式诱导细胞死亡并降低PDAC细胞的活力和克隆形成性。此外,NPS-1034通过抑制MET/PI3K/AKT轴诱导的上皮-间质转化(EMT)来抑制PDAC细胞的迁移。NPS-1034与氟尿嘧啶或奥沙利铂的组合显示出协同作用,与单一疗法相比,显着降低细胞活力并诱导肿瘤细胞凋亡。下一代测序提供的机制见解表明,NPS-1034通过诱导PDAC细胞中I型干扰素和肿瘤坏死因子的产生来调节免疫应答。这表明NPS-1034除了MET和AXL抑制外还有更广泛的作用,将其定位为潜在的免疫调节剂。总的来说,这些发现强调了NPS-1034在体外PDAC治疗中的抗癌潜力,无论是作为单一疗法还是与传统化疗相结合,为临床探索之前的进一步体内研究提供了一个有希望的途径。
    Pancreatic ductal adenocarcinoma (PDAC) poses a significant challenge in terms of diagnosis and treatment, with limited therapeutic options and a poor prognosis. This study explored the potential therapeutic role of NPS-1034, a kinase inhibitor targeting MET and AXL, in PDAC. The investigation included monotherapy with NPS-1034 and its combination with the commonly prescribed chemotherapy agents, fluorouracil and oxaliplatin. Our study revealed that NPS-1034 induces cell death and reduces the viability and clonogenicity of PDAC cells in a dose-dependent manner. Furthermore, NPS-1034 inhibits the migration of PDAC cells by suppressing MET/PI3K/AKT axis-induced epithelial-to-mesenchymal transition (EMT). The combination of NPS-1034 with fluorouracil or oxaliplatin demonstrated a synergistic effect, significantly reducing cell viability and inducing tumor cell apoptosis compared to monotherapies. Mechanistic insights provided by next-generation sequencing indicated that NPS-1034 modulates immune responses by inducing type I interferon and tumor necrosis factor production in PDAC cells. This suggests a broader role for NPS-1034 beyond MET and AXL inhibition, positioning it as a potential immunity modulator. Overall, these findings highlight the anticancer potential of NPS-1034 in PDAC treatment in vitro, both as a monotherapy and in combination with traditional chemotherapy, offering a promising avenue for further in vivo investigation before clinical exploration.
    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
    吉西他滨是一种广泛使用的嘧啶结构的抗代谢药物,它可以作为游离碱分子形式(Gem)存在。药物的包封形式对于延迟和局部药物释放是令人感兴趣的。我们利用,第一次,一种通过液体辅助研磨(LAG)将宝石封装在卟啉铝金属有机骨架Al-MOF-TCPPH2(化合物2)的“基质”上的机械化学新方法。通过ATR-FTIR光谱和粉末XRD研究了宝石与化合物2的化学键合。这种相互作用涉及宝石分子的C=O基团,这表明在获得的复合物中形成了包封复合物。Further,使用配备有自动进样器的自动药物溶解装置,在37°C下研究Gem在磷酸盐缓冲盐水(PBS)中的延迟释放。通过HPLC-UV分析确定释放的药物的浓度。由于结合形式和此后的恒定浓度,该复合材料显示出Gem的延迟释放,而纯Gem显示在小于45分钟内快速溶解。Gem药物从复合材料中的延迟释放遵循动力学假一级速率定律。Further,第一次,通过药物释放介质的可变搅拌速度来评估Gem的延迟释放机理,当搅拌速度降低(扩散控制)时,发现动力学速率常数k降低。最后,通过连续测量增殖(生长)6天,研究了Gem对胰腺癌PANC-1细胞毒性的延长时间尺度,使用xCELLigence实时细胞分析仪(RTCA),对于复合材料与纯药物,它们的差异表明药物释放延迟。铝金属-有机骨架是用于封装吉西他滨和相关小分子抗代谢物的新的有前途的材料,用于控制药物延迟释放和在药物洗脱植入物中的潜在用途。
    Gemcitabine is a widely used antimetabolite drug of pyrimidine structure, which can exist as a free-base molecular form (Gem). The encapsulated forms of medicinal drugs are of interest for delayed and local drug release. We utilized, for the first time, a novel approach of mechano-chemistry by liquid-assisted grinding (LAG) to encapsulate Gem on a \"matrix\" of porphyrin aluminum metal-organic framework Al-MOF-TCPPH2 (compound 2). The chemical bonding of Gem to compound 2 was studied by ATR-FTIR spectroscopy and powder XRD. The interaction involves the C=O group of Gem molecules, which indicates the formation of the encapsulation complex in the obtained composite. Further, the delayed release of Gem from the composite was studied to phosphate buffered saline (PBS) at 37 °C using an automated drug dissolution apparatus equipped with an autosampler. The concentration of the released drug was determined by HPLC-UV analysis. The composite shows delayed release of Gem due to the bonded form and constant concentration thereafter, while pure Gem shows quick dissolution in less than 45 min. Delayed release of Gem drug from the composite follows the kinetic pseudo-first-order rate law. Further, for the first time, the mechanism of delayed release of Gem was assessed by the variable stirring speed of drug release media, and kinetic rate constant k was found to decrease when stirring speed is decreased (diffusion control). Finally, the prolonged time scale of toxicity of Gem to pancreatic cancer PANC-1 cells was studied by continuous measurements of proliferation (growth) for 6 days, using the xCELLigence real-time cell analyzer (RTCA), for the composite vs. pure drug, and their differences indicate delayed drug release. Aluminum metal-organic frameworks are new and promising materials for the encapsulation of gemcitabine and related small-molecule antimetabolites for controlled delayed drug release and potential use in drug-eluting implants.
    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
    胰腺腺癌(PAAD)的早期检测仍然是一个紧迫的临床问题。关于PAAD中线粒体融合相关基因的临床预后价值的信息仍然有限。在这项研究中,我们研究了PAAD的线粒体融合相关基因,以建立PAAD的早期诊断和预后的最佳特征板。癌症基因组图谱数据库用于整合PAAD患者的每千碱基百万片段数据和相关临床数据。最小绝对收缩和选择算子回归,cox回归,工作特性曲线,并使用cBioPortal数据库评估模型性能,评估预后能力和敏感性。通过CIBERSORT比较免疫浸润水平,QUANTISER,和EPIC。通过癌症药物敏感性基因组学数据库和“pRophetic”R软件包比较了不同风险组之间的化疗敏感性。最后,共有4个基因纳入多变量Cox回归分析.风险预测特征构建为:(0.5438×BAK1)(-1.0259×MIGA2)(1.1140×PARL)(-0.4300×PLD6)。这4个基因的曲线下面积为0.89。Cox回归分析表明该特征是独立的预后指标(P<.001,风险比[HR]=1.870,95%CI=1.568-2.232)。使用3种算法观察2个风险组中不同程度的免疫细胞浸润,与肿瘤突变负荷(P=.0063),肿瘤微环境评分(P=0.01),和肿瘤免疫功能障碍和排斥评分(P=.0012)。化疗敏感性分析还显示,5-氟尿嘧啶的半数最大抑制浓度(P=0.0127),顺铂(P=0.0099),多西他赛(P<0.0001),吉西他滨(P=.0047),和帕拉他赛(P<0.0001)在高风险组中较低,表明高危组患者对化疗的敏感性更高。总之,我们建立了由4个线粒体融合相关基因组成的基因标签板,以促进PAAD的早期诊断和预后预测。
    Early detection of pancreatic adenocarcinoma (PAAD) remains a pressing clinical problem. Information on the clinical prognostic value of mitochondrial fusion-related genes in PAAD remains limited. In this study, we investigated mitochondrial fusion-related genes of PAAD to establish an optimal signature plate for the early diagnosis and prognosis of PAAD. The cancer genome atlas database was used to integrate the Fragments Per Kilobase Million data and related clinical data for patients with PAAD. Least absolute shrinkage and selection operator regression, cox regression, operating characteristic curves, and cBioPortal database was used to evaluate model performance, assess the prognostic ability and sensitivity. The levels of immune infiltration were compared by CIBERSORT, QUANTISEQ, and EPIC. Chemotherapy sensitivity between the different risk groups was compared by the Genomics of Drug Sensitivity in Cancer database and the \"pRRophetic\" R package. At last, a total of 4 genes were enrolled in multivariate Cox regression analysis. The risk-predictive signature was constructed as: (0.5438 × BAK1) + (-1.0259 × MIGA2) + (1.1140 × PARL) + (-0.4300 × PLD6). The area under curve of these 4 genes was 0.89. Cox regression analyses indicates the signature was an independent prognostic indicator (P < .001, hazard ratio [HR] = 1.870, 95% CI = 1.568-2.232). Different levels of immune cell infiltration in the 2 risk groups were observed using the 3 algorithms, with tumor mutation load (P = .0063), tumor microenvironment score (P = .01), and Tumor Immune Dysfunction and Exclusion score (P = .0012). The chemotherapeutic sensitivity analysis also revealed that the half-maximal inhibitory concentration of 5-fluorouracil (P = .0127), cisplatin (P = .0099), docetaxel (P < .0001), gemcitabine (P = .0047), and pacilataxel (P < .0001) were lower in the high-risk groups, indicating that the high-risk group patients had a greater sensitivity to chemotherapy. In conclude, we established a gene signature plate comprised of 4 mitochondrial fusion related genes to facilitate early diagnosis and prognostic prediction of PAAD.
    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
    细胞外HSP90α(eHSP90α)是肿瘤发展和恶性进展的启动子。恶性肿瘤患者,包括胰腺导管腺癌(PDAC),通常显示血清/血浆eHSP90α水平增加5~10倍。在这项研究中,我们开发了针对eHSP90α的人源化抗体HH01,并评估了其抗癌功效。HH01,具有新的互补决定区,表现出对HSP90α的高结合亲和力。它识别HSP90α表位位点235AEEKEDKEEE244和251ESEDKPEIED260,具有关键氨基酸残基E237,E239,D240,K241,E253和K255。HH01通过阻断eHSP90α与细胞表面受体CD91的连接,有效抑制eHSP90α诱导的结直肠癌和PDAC细胞系的侵袭性和球状体形成活性。在老鼠模型中,HH01有效抑制由内皮-间质转化衍生的癌症相关成纤维细胞促进的PDAC细胞移植物/异种移植物的肿瘤生长,同时还降低血清eHSP90α水平,反映了其抗癌功效。HH01还通过减少M2巨噬细胞和恢复免疫T细胞来调节肿瘤免疫。此外,HH01显示低聚集倾向,高水溶性,在小鼠血液中的半衰期>18天。对视网膜色素上皮细胞无细胞毒性,对小鼠器官无明显毒性。我们的数据表明,用HH01抗体靶向eHSP90α可能是PDAC治疗的有希望的新策略。
    Extracellular HSP90α (eHSP90α) is a promoter of tumor development and malignant progression. Patients with malignancies, including pancreatic ductal adenocarcinoma (PDAC), have generally shown 5~10-fold increases in serum/plasma eHSP90α levels. In this study, we developed a humanized antibody HH01 to target eHSP90α and evaluated its anticancer efficacy. HH01, with novel complementarity-determining regions, exhibits high binding affinity toward HSP90α. It recognizes HSP90α epitope sites 235AEEKEDKEEE244 and 251ESEDKPEIED260, with critical amino acid residues E237, E239, D240, K241, E253, and K255. HH01 effectively suppressed eHSP90α-induced invasive and spheroid-forming activities of colorectal cancer and PDAC cell lines by blocking eHSP90α\'s ligation with the cell-surface receptor CD91. In mouse models, HH01 potently inhibited the tumor growth of PDAC cell grafts/xenografts promoted by endothelial-mesenchymal transition-derived cancer-associated fibroblasts while also reducing serum eHSP90α levels, reflecting its anticancer efficacy. HH01 also modulated tumor immunity by reducing M2 macrophages and reinvigorating immune T-cells. Additionally, HH01 showed low aggregation propensity, high water solubility, and a half-life time of >18 days in mouse blood. It was not cytotoxic to retinal pigmented epithelial cells and showed no obvious toxicity in mouse organs. Our data suggest that targeting eHSP90α with HH01 antibody can be a promising novel strategy for PDAC therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号