cancer stem cell

肿瘤干细胞
  • 文章类型: Journal Article
    癌症进展涉及分化表型的逐渐丧失和祖细胞和干细胞样特征的获得。是免疫疗法抵抗的潜在元凶。尽管最先进的预测计算方法促进了癌症干性的预测,目前还没有能够满足各种使用需求的高效资源。这里,我们在网上展示癌症干细胞,在批量和单细胞水平上有效评估癌症干细胞潜能的综合资源。该资源集成了8个强大的预测算法以及与癌症干性相关的27个签名基因集,用于预测干性评分。从五个不同的方面进行了下游分析,包括确定癌症干性的特征基因,探索与癌症标志的关联,细胞状态,免疫反应,以及与免疫细胞的交流;调查对患者生存的贡献;并在不同方法中对癌症干性进行稳健性分析。此外,用户可以访问40多种癌症类型的预先计算的癌症干性图谱。可以下载分析结果的表格和各种可视化。一起,癌症干细胞在线是癌症干细胞评分和扩展下游功能解释的强大资源,包括免疫反应以及癌症标志。癌症干细胞在线可以在http://bio-bigdata上免费访问。hrbmu.edu.cn/CancerStemnessOnline.
    Cancer progression involves the gradual loss of a differentiated phenotype and the acquisition of progenitor and stem-cell-like features, which are potential culprits of immunotherapy resistance. Although the state-of-art predictive computational methods have facilitated the prediction of cancer stemness, currently there is no efficient resource that can meet various usage requirements. Here, we present the Cancer Stemness Online, an integrated resource for efficiently scoring cancer stemness potential at the bulk and single-cell levels. The resource integrates 8 robust predictive algorithms as well as 27 signature gene sets associated with cancer stemness for predicting stemness scores. Downstream analyses were performed from five different aspects, including identifying the signature genes of cancer stemness, exploring the associations with cancer hallmarks, cellular states, the immune response, and communication with immune cells; investigating the contributions to patient survival; and performing a robustness analysis of cancer stemness among different methods. Moreover, the pre-calculated cancer stemness atlas for more than 40 cancer types can be accessed by users. Both the tables and diverse visualizations of the analytical results are available for download. Together, Cancer Stemness Online is a powerful resource for scoring cancer stemness and expanding the downstream functional interpretation, including immune response as well as cancer hallmarks. Cancer Stemness Online is freely accessible at http://bio-bigdata.hrbmu.edu.cn/CancerStemnessOnline.
    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:癌症相关成纤维细胞(CAF)是肿瘤微环境(TME)中突出的细胞类型,已经在各种肿瘤中鉴定了CAF亚群。然而,CAF如何在空间上协调肝脏TME内的其他细胞群体以促进癌症进展尚不清楚.
    方法:我们结合了多区域蛋白质组学(6例患者,24个样本),10X基因组学空间转录组学(11名患者,25个样品),和多重成像(92名患者,264个样本)破译表达异质性的技术,功能多样性,空间分布,共同定位,和成纤维细胞的相互作用。通过从5名肝癌患者中分离的细胞和体外功能测定来验证新鉴定的CAF亚群。
    结果:我们确定了肝脏CAF亚群,以COL1A2、COL4A1、COL4A2、CTGF、和FSTL1,并命名为F5-CAF。F5-CAF优先位于肿瘤巢内和周围,并与肝细胞癌(HCC)中具有较高干性的癌细胞共定位。92例患者的多重染色和371例患者的大量转录组表明,HCC中F5-CAFs的丰度与预后较差有关。进一步的体外实验表明,从肝癌患者中分离的F5-CAFs可以促进HCC细胞的增殖和干性。
    结论:我们确定了肝癌中的CAF亚群F5-CAF,这与癌症的干性和不良预后有关。我们的结果提供了TME中CAF亚群通过支持癌症干细胞的存活来促进肝癌发展的潜在机制。
    BACKGROUND: Cancer-associated fibroblasts (CAFs) are the prominent cell type in the tumor microenvironment (TME), and CAF subsets have been identified in various tumors. However, how CAFs spatially coordinate other cell populations within the liver TME to promote cancer progression remains unclear.
    METHODS: We combined multi-region proteomics (6 patients, 24 samples), 10X Genomics Visium spatial transcriptomics (11 patients, 25 samples), and multiplexed imaging (92 patients, 264 samples) technologies to decipher the expression heterogeneity, functional diversity, spatial distribution, colocalization, and interaction of fibroblasts. The newly identified CAF subpopulation was validated by cells isolated from 5 liver cancer patients and in vitro functional assays.
    RESULTS: We identified a liver CAF subpopulation, marked by the expression of COL1A2, COL4A1, COL4A2, CTGF, and FSTL1, and named F5-CAF. F5-CAF is preferentially located within and around tumor nests and colocalizes with cancer cells with higher stemness in hepatocellular carcinoma (HCC). Multiplexed staining of 92 patients and the bulk transcriptome of 371 patients demonstrated that the abundance of F5-CAFs in HCC was associated with a worse prognosis. Further in vitro experiments showed that F5-CAFs isolated from liver cancer patients can promote the proliferation and stemness of HCC cells.
    CONCLUSIONS: We identified a CAF subpopulation F5-CAF in liver cancer, which is associated with cancer stemness and unfavorable prognosis. Our results provide potential mechanisms by which the CAF subset in the TME promotes the development of liver cancer by supporting the survival of cancer stem cells.
    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
    索洛酮酰胺是半合成的三萜类化合物,可以在体外和体内穿过血脑屏障并抑制胶质母细胞瘤的生长。在这里,我们研究了这些化合物对胶质母细胞瘤侵袭性和治疗抗性相关过程的影响。针对胶质母细胞瘤细胞的索洛酮酰胺的筛选揭示了化合物7(索洛酮对甲基苯胺)抑制转化生长因子β1(TGF-β1)诱导的神经胶质间质转化的能力化合物7抑制了形态学变化,伤口愈合,Transwell迁移,和间充质标志物(N-cadherin,纤连蛋白,Slug)在TGF-β1诱导的U87和U118胶质母细胞瘤细胞中,同时恢复它们的粘附性。共聚焦显微镜和分子对接显示,7可能通过与TGF-βI型和II型受体(TβRI/II)的直接相互作用来减少SMAD2/3核易位。此外,7抑制胶质母细胞瘤细胞的干性,如集落形成能力的抑制所证明,球体生长,和醛脱氢酶(ALDH)活性。此外,图7显示了与替莫唑胺(TMZ)对成胶质细胞瘤细胞活力的协同作用。使用N-乙酰-L-半胱氨酸(NAC)和流式细胞术分析膜联蛋白V-FITC-,碘化丙啶-,和DCFDA染色的细胞,发现7通过诱导ROS依赖性凋亡协同TMZ的细胞毒性。进一步的体内研究表明,7,单独或与TMZ联合使用,有效抑制小鼠U87异种移植瘤的生长。因此,7证明了作为胶质母细胞瘤联合治疗的组成部分的有希望的潜力,降低其侵袭性并增加其对化疗的敏感性。
    Soloxolone amides are semisynthetic triterpenoids that can cross the blood-brain barrier and inhibit glioblastoma growth both in vitro and in vivo. Here we investigate the impact of these compounds on processes associated with glioblastoma invasiveness and therapy resistance. Screening of soloxolone amides against glioblastoma cells revealed the ability of compound 7 (soloxolone para-methylanilide) to inhibit transforming growth factor-beta 1 (TGF-β1)-induced glial-mesenchymal transition Compound 7 inhibited morphological changes, wound healing, transwell migration, and expression of mesenchymal markers (N-cadherin, fibronectin, Slug) in TGF-β1-induced U87 and U118 glioblastoma cells, while restoring their adhesiveness. Confocal microscopy and molecular docking showed that 7 reduced SMAD2/3 nuclear translocation probably by direct interaction with the TGF-β type I and type II receptors (TβRI/II). In addition, 7 suppressed stemness of glioblastoma cells as evidenced by inhibition of colony forming ability, spheroid growth, and aldehyde dehydrogenase (ALDH) activity. Furthermore, 7 exhibited a synergistic effect with temozolomide (TMZ) on glioblastoma cell viability. Using N-acetyl-L-cysteine (NAC) and flow cytometry analysis of Annexin V-FITC-, propidium iodide-, and DCFDA-stained cells, 7 was found to synergize the cytotoxicity of TMZ by inducing ROS-dependent apoptosis. Further in vivo studies showed that 7, alone or in combination with TMZ, effectively suppressed the growth of U87 xenograft tumors in mice. Thus, 7 demonstrated promising potential as a component of combination therapy for glioblastoma, reducing its invasiveness and increasing its sensitivity to chemotherapy.
    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:本研究旨在研究癌症干细胞(CSC)相关基因的综合表达谱,并构建高风险肾母细胞瘤(WT)的总体生存(OS)预测的预后标志。材料和方法:使用来自产生有效治疗的治疗应用研究(TARGET)-WT中的120个高风险WT病例的基因表达和存活数据。结果:总的来说,与肿瘤附近的正常组织相比,在WT中发现229个CSC相关基因显着失调,其中34个与OS相关。使用LASSO回归,开发了22个基因签名,在3-,5-,和10年操作系统预测(AUC>0.86)。与低风险评分组相比,高风险评分组显示出明显较差的OS(中位数分离,HR=6.41,95%CI:3.18-12.92,p=3.2e-9)。22基因标记是OS的独立预后因素(HR=5.086,95%CI:3.019-8.568,p<0.001)。结论:这项研究确定了一个强大的预后特征,可以有效地支持OS预测。
    Background: This study aimed to investigate the comprehensive expression profile of cancer stem cell (CSC)-related genes and construct a prognostic signature for overall survival (OS) prediction in high-risk Wilms\' tumor (WT). Materials and methods: Gene expression and survival data from 120 high-risk WT cases in the Therapeutically Applicable Research to Generate Effective Treatments (TARGET)-WT were used. Results: In total, 229 CSC-related genes were found to be significantly dysregulated in WT compared to tumor-adjacent normal tissues, among which 34 were associated with OS. Using LASSO regression, a 22-gene signature was developed, which exhibited excellent performance in 3-, 5-, and 10-year OS predictions (AUC > 0.86). The high-risk score group showed markedly poorer OS compared to the low-risk score group (median separation, HR = 6.41, 95% CI: 3.18-12.92, p = 3.2e - 9). The 22-gene signature was an independent prognostic factor for OS (HR = 5.086, 95% CI: 3.019-8.568, p < 0.001). Conclusion: This study identified a robust prognostic signature that can effectively support OS prediction.
    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
    肿瘤干细胞(CSC)构成了肿瘤微环境(TME)中的关键元素,驱动癌症的开始和进展。然而,喉鳞状细胞癌(LSCC)中CSC的鉴定及其潜在的分子机制仍然是一项艰巨的挑战.我们采用了匹配的原发性肿瘤组织的单细胞RNA测序,癌旁组织,和3例LSCC患者的局部淋巴结。来自上皮群体的干细胞的两个不同簇被描绘和验证为CSC和正常干细胞(NSC)。分别。与肿瘤组织相比,CSC在癌旁组织中含量丰富。CSCs高表达干细胞标记基因如PROM1、ALDH1A1、SOX4,并增加肿瘤相关缺氧的活性,Wnt/β-catenin,和Notch信号通路。然后,我们探索了CSC和TME细胞之间的复杂串扰,并确定了TME中与CSC相关的靶标。我们还发现了8个与LSCC患者预后显著相关的CSCs标记基因。此外,生物信息学分析表明,厄洛替尼等药物,OSI-027和依鲁替尼选择性靶向CSC特异性表达的基因。总之,我们的结果代表了在单细胞水平上对LSCC中CSC特性的首次全面表征。
    Cancer stem cells (CSCs) constitute a pivotal element within the tumor microenvironment (TME), driving the initiation and progression of cancer. However, the identification of CSCs and their underlying molecular mechanisms in laryngeal squamous cell carcinoma (LSCC) remains a formidable challenge. We employed single-cell RNA sequencing of matched primary tumor tissues, paracancerous tissues, and local lymph nodes from three LSCC patients. Two distinct clusters of stem cells originating from epithelial populations were delineated and verified as CSCs and normal stem cells (NSCs), respectively. CSCs were abundant in the paracancerous tissues compared to the tumor tissues. CSCs showed high expression of stem cell marker genes such as PROM1, ALDH1A1, and SOX4, and increased the activity of tumor-related hypoxia, Wnt/β-catenin, and Notch signaling pathways. We then explored the intricate crosstalk between CSCs and the TME cells and identified targets within the TME that related with CSCs. We also found eight marker genes of CSCs that correlated significantly with the prognosis of LSCC patients. Furthermore, bioinformatics analyses showed that drugs such as erlotinib, OSI-027, and ibrutinib selectively targeted the CSC-specifically expressed genes. In conclusion, our results represent the first comprehensive characterization of CSCs properties in LSCC at the single-cell level.
    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

    高转移,对普通治疗的抵抗力,和高死亡率,使三阴性乳腺癌(TNBC)成为最具侵袭性的乳腺癌类型。高端粒酶活性和线粒体生物发生参与乳腺癌的发生。端粒酶的催化亚基,端粒酶逆转录酶(hTERT),在端粒延长和基因表达等生物学外功能中起作用,线粒体功能,和凋亡。在这项研究中,它的目的是评估内在的-,在TNBC细胞系中抑制端粒酶和线粒体呼吸后,外源性凋亡以及DNMT3a和TET2的表达。
    TNBC细胞用IC50水平的BIBR1532,替加环素,还有他们的组合。然后,端粒长度,和DNMT3a,评估TET2和hTERT表达。最后,凋亡率,凋亡相关蛋白,并对基因进行了分析。
    目前的结果表明,端粒酶的IC50水平和线粒体呼吸的抑制诱导了细胞凋亡,但对端粒长度没有任何显着影响。结果还表明,端粒酶抑制在MDA-MB-231细胞中诱导外源性凋亡,并在MDA-MB-468细胞中引起内源性凋亡。此外,发现p53的表达降低,对细胞凋亡无效。细胞中DNMT3a和TET2的表达增加。此外,联合治疗优于BIBR1532和替加环素单独治疗。
    端粒酶和线粒体呼吸的抑制可引起体内和外凋亡,并增加DNMT3a和TET2的表达,可用于乳腺癌的治疗。

    UNASSIGNED: High metastasis, resistance to common treatments, and high mortality rate, has made triple-negative breast cancer (TNBC) to be the most invasive type of breast cancer. High telomerase activity and mitochondrial biogenesis are involved in breast cancer tumorigenesis. The catalytic subunit of telomerase, telomerase reverse transcriptase (hTERT), plays a role in telomere lengthening and extra-biological functions such as gene expression, mitochondria function, and apoptosis. In this study, it has been aimed to evaluate intrinsic-, extrinsic-apoptosis and DNMT3a and TET2 expression following the inhibition of telomerase and mitochondria respiration in TNBC cell lines.
    UNASSIGNED: TNBC cells were treated with IC50 levels of BIBR1532, tigecycline, and also their combination. Then, telomere length, and DNMT3a, TET2, and hTERT expression were evaluated. Finally, apoptosis rate, apoptosis-related proteins, and genes were analyzed.
    UNASSIGNED: The present results showed that IC50 level of telomerase and inhibition of mitochondria respiration induced apoptosis but did not leave any significant effect on telomere length. The results also indicated that telomerase inhibition induced extrinsic-apoptosis in MDA-MB-231 and caused intrinsic- apoptosis in MDA-MB-468 cells. Furthermore, it was found that the expression of p53 decreased and was ineffective in cell apoptosis. The expressions of DNMT3a and TET2 increased in cells. In addition, combination treatment was better than BIBR1532 and tigecycline alone.
    UNASSIGNED: The inhibition of telomerase and mitochondria respiration caused intrinsic- and extrinsic- apoptosis and increased DNMT3a and TET2 expression and it could be utilized in breast cancer treatment.
    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
    免疫原性细胞死亡(ICD)可以激活抗肿瘤免疫反应,这对于提高癌症治疗效果非常有吸引力。这里,这项工作报道了一种多功能砷(III)变构抑制剂Mech02,它通过敏化的生物催化反应诱导1O2的过度积累,导致细胞焦亡和放大的ICD效应。Mech02转化为Mech03后,对丙酮酸激酶M2的变构口袋产生更强的结合作用,进一步干扰肿瘤的无氧糖酵解途径。由Mech02引发的增强的DNA损伤和癌症干细胞的焦亡为完全清除肿瘤提供了保证。体内实验证明纳米粒Mech02-HANP能够激活免疫记忆效应并提高抗肿瘤免疫的持久性。总之,本研究首次将砷(Ⅲ)药效团作为增强ICD效应引发剂引入氮芥,为开发有效的多模式肿瘤治疗剂提供见解。
    Immunogenic cell death (ICD) could activate anti-tumor immune responses, which is highly attractive for improving cancer treatment effectiveness. Here, this work reports a multifunctional arsenic(III) allosteric inhibitor Mech02, which induces excessive accumulation of 1O2 through sensitized biocatalytic reactions, leading to cell pyroptosis and amplified ICD effect. After Mech02 is converted to Mech03, it could actualize stronger binding effects on the allosteric pocket of pyruvate kinase M2, further interfering with the anaerobic glycolysis pathway of tumors. The enhanced DNA damage triggered by Mech02 and the pyroptosis of cancer stem cells provide assurance for complete tumor clearance. In vivo experiments prove nanomicelle Mech02-HA NPs is able to activate immune memory effects and raise the persistence of anti-tumor immunity. In summary, this study for the first time to introduce the arsenic(III) pharmacophore as an enhanced ICD effect initiator into nitrogen mustard, providing insights for the development of efficient multimodal tumor therapy agents.
    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
    干细胞的标志,如扩散,自我更新,发展,分化,和再生,对于维持由遗传和表观遗传因素维持的干细胞身份至关重要。超级增强剂(SE),由活性增强剂簇组成,通过特定的转录模型在维持干性标志方面发挥核心作用。SE导航转录复合物,包括SE,非编码RNA,掌握转录因子,调解员和其他共同激活者,形成相分离的冷凝物,它提供了指导不同干细胞命运的切换。随着多元组学技术的蓬勃发展,应用于研究SE的不同方面,我们首先提出了“超级增强子组学”的概念,与泛经济学有着千丝万缕的联系。在审查中,我们讨论了SE的时空组织和概念,并描述SE导航转录复合物和干细胞特征之间的联系,比如干细胞身份,自我更新,多能性,分化和发展。我们还阐明了通过癌症干细胞中的基因组和表观遗传改变劫持来调节癌症干细胞的干性和致癌SEs的机制。此外,我们讨论了使用小分子化合物靶向SE复合物成分的潜力,基因组编辑,和反义寡核苷酸治疗SE相关的器官功能障碍和疾病,包括癌症.这篇综述还通过SE的范式提供了对干细胞研究未来的见解。
    The hallmarks of stem cells, such as proliferation, self-renewal, development, differentiation, and regeneration, are critical to maintain stem cell identity which is sustained by genetic and epigenetic factors. Super-enhancers (SEs), which consist of clusters of active enhancers, play a central role in maintaining stemness hallmarks by specifically transcriptional model. The SE-navigated transcriptional complex, including SEs, non-coding RNAs, master transcriptional factors, Mediators and other co-activators, forms phase-separated condensates, which offers a toggle for directing diverse stem cell fate. With the burgeoning technologies of multiple-omics applied to examine different aspects of SE, we firstly raise the concept of \"super-enhancer omics\", inextricably linking to Pan-omics. In the review, we discuss the spatiotemporal organization and concepts of SEs, and describe links between SE-navigated transcriptional complex and stem cell features, such as stem cell identity, self-renewal, pluripotency, differentiation and development. We also elucidate the mechanism of stemness and oncogenic SEs modulating cancer stem cells via genomic and epigenetic alterations hijack in cancer stem cell. Additionally, we discuss the potential of targeting components of the SE complex using small molecule compounds, genome editing, and antisense oligonucleotides to treat SE-associated organ dysfunction and diseases, including cancer. This review also provides insights into the future of stem cell research through the paradigm of SEs.
    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
    肿瘤干细胞(CSC)引起的频繁复发和转移是肺癌治疗的主要挑战。因此,确定和表征特定CSC靶标对于前瞻性靶向治疗的成功至关重要.在这项研究中,发现肺CSC中上调的TOR信号通路调节因子样(TIPRL)通过与CaMKK2结合,导致钙/钙调蛋白依赖性蛋白激酶激酶2(CaMKK2)信号通路的持续激活,从而维持干细胞和存活。CaMKK2介导的CaM激酶4(CaMK4)的激活导致cAMP反应元件结合蛋白(CREB)在Ser129和Ser133处的磷酸化,这对于其最大激活和其靶基因(Bcl2和HMG20A)的下游组成型表达是必需的。TIPRL耗竭使肺CSC对阿法替尼诱导的细胞死亡敏感,并减少体内肺癌的远端转移。确定CREB激活肺CSC中TIPRL的转录。由CREB和TIPRL组成的正反馈回路诱导作为驱动力的CaMKK2-CaMK4-CREB轴的持续激活,并上调干细胞和存活相关基因的表达,促进肺癌患者的肿瘤发生。因此,TIPRL和CaMKK2信号轴可能是克服肺癌耐药性和减少转移的有希望的靶标。
    Frequent recurrence and metastasis caused by cancer stem cells (CSCs) are major challenges in lung cancer treatment. Therefore, identifying and characterizing specific CSC targets are crucial for the success of prospective targeted therapies. In this study, it is found that upregulated TOR Signaling Pathway Regulator-Like (TIPRL) in lung CSCs causes sustained activation of the calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2) signaling pathway by binding to CaMKK2, thereby maintaining stemness and survival. CaMKK2-mediated activation of CaM kinase 4 (CaMK4) leads to phosphorylation of cAMP response element-binding protein (CREB) at Ser129 and Ser133, which is necessary for its maximum activation and the downstream constitutive expression of its target genes (Bcl2 and HMG20A). TIPRL depletion sensitizes lung CSCs to afatinib-induced cell death and reduces distal metastasis of lung cancer in vivo. It is determined that CREB activates the transcription of TIPRL in lung CSCs. The positive feedback loop consisting of CREB and TIPRL induces the sustained activation of the CaMKK2-CaMK4-CREB axis as a driving force and upregulates the expression of stemness- and survival-related genes, promoting tumorigenesis in patients with lung cancer. Thus, TIPRL and the CaMKK2 signaling axis may be promising targets for overcoming drug resistance and reducing metastasis in lung 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
    盐霉素(Sal)在肿瘤治疗领域引起了广泛的关注,尤其是对肿瘤干细胞(CSCs)和耐药肿瘤细胞的抑制作用。然而,其溶解性和靶向特异性对其药物开发提出了重大挑战。Sal-A6,一种新型的肽-药物偶联物(PDC),通过使用特异性接头将靶向CSC标记CD44的肽A6与Sal连接而形成。这种共轭显着增强了Sal的物理化学性质,与Sal相比,Sal-A6表现出显著增加的抗卵巢癌活性。此外,Sal-A6,使用二硫键作为接头,表现出旁观者的杀戮效果。此外,除了增强耐药卵巢癌细胞的化学敏感性外,它还对癌症干细胞和耐药细胞具有实质性的细胞毒性作用。总之,结果表明,Sal-A6是一种衍生自Sal的新型PDC,在卵巢癌和耐药患者的治疗中具有潜在的治疗应用。此外,这一发现为以Sal为基础开发PDC型药物提供了见解.
    Salinomycin (Sal) has attracted considerable attention in the field of tumor treatment, especially for its inhibitory effect on cancer stem cells (CSCs) and drug-resistant tumor cells. However, its solubility and targeting specificity pose significant challenges to its pharmaceutical development. Sal-A6, a novel peptide-drug conjugate (PDC), was formed by linking the peptide A6 targeting the CSC marker CD44 with Sal using a specific linker. This conjugation markedly enhances the physicochemical properties of Sal and compared to Sal, Sal-A6 demonstrated a significantly increased activity against ovarian cancer. Furthermore, Sal-A6, employing a disulfide bond as a linker, exhibited bystander killing effect. Moreover, it induces substantial cytotoxic effect on both cancer stem cells and drug-resistant cells in addition to enhance chemosensitivity of resistant ovarian cancer cells. In summary, the results indicated that Sal-A6, a novel PDC derived from Sal, has potential therapeutic applications in the treatment of ovarian cancer and drug-resistant patients. Additionally, this discovery offers insights for developing PDC-type drugs using Sal as a foundation.
    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