Mesothelin

间皮素
  • 文章类型: Journal Article
    卵巢癌是最致命的妇科恶性肿瘤,占所有妇科癌症相关死亡的近65%。早期检测和诊断的挑战,再加上癌细胞广泛的腹膜内扩散和对化疗的耐药性,这种疾病的高死亡率。由于缺乏特定的症状和缺乏有效的筛查方法,大多数卵巢癌病例被诊断为晚期。虽然化疗是一种常见的治疗方法,它经常导致肿瘤复发,需要进一步的干预。近年来,抗体-药物偶联物(ADC)已成为靶向癌症治疗中一种有价值的工具.这些复杂的生物治疗剂通过接头将特异性靶向肿瘤特异性/相关抗原的抗体与高效抗癌药物相结合。为卵巢癌的治疗提供了一种有希望的方法。各种人类肿瘤中分子靶标的鉴定为靶向治疗的发展铺平了道路。ADC处于这一创新的最前沿。通过将细胞毒性剂直接递送至肿瘤和转移性病变,ADC显示出管理化学抗性卵巢癌的潜力。与正常和/或良性样品相比,粘蛋白如MUC16、MUC13和MUC1在卵巢肿瘤中显示出显著更高的表达,因此已成为ADC生成的有希望的目标。虽然传统标志物在非癌性疾病中受其升高水平的限制,粘蛋白为卵巢癌的靶向治疗提供了新的可能性。这篇综述全面描述了粘蛋白在ADC治疗产生中的潜力,强调它们在寻求改善卵巢癌患者预后方面的重要性。
    Ovarian cancer stands as the deadliest gynecologic malignancy, responsible for nearly 65% of all gynecologic cancer-related deaths. The challenges in early detection and diagnosis, coupled with the widespread intraperitoneal spread of cancer cells and resistance to chemotherapy, contribute significantly to the high mortality rate of this disease. Due to the absence of specific symptoms and the lack of effective screening methods, most ovarian cancer cases are diagnosed at advanced stages. While chemotherapy is a common treatment, it often leads to tumor recurrence, necessitating further interventions. In recent years, antibody-drug conjugates (ADCs) have emerged as a valuable tool in targeted cancer therapy. These complex biotherapeutics combine an antibody that specifically targets tumor specific/associated antigen(s) with a high potency anti-cancer drug through a linker, offering a promising approach for ovarian cancer treatment. The identification of molecular targets in various human tumors has paved the way for the development of targeted therapies, with ADCs being at the forefront of this innovation. By delivering cytotoxic agents directly to tumors and metastatic lesions, ADCs show potential in managing chemo-resistant ovarian cancers. Mucins such as MUC16, MUC13, and MUC1 have shown significantly higher expression in ovarian tumors as compared to normal and/or benign samples, thus have become promising targets for ADC generation. While traditional markers are limited by their elevated levels in non-cancerous conditions, mucins offer a new possibility for targeted treatment in ovarian cancer. This review comprehensively described the potential of mucins for the generation of ADC therapy, highlighting their importance in the quest to improve the outcome of ovarian cancer patients.
    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
    In 1993, a total asbestos ban was introduced in Germany. Thirty years later, mesothelioma is still one of the most frequent occupational diseases. Recent data on incidence, mortality, recognized occupational diseases, early detection, and assessment are presented in this article.
    UNASSIGNED: In Deutschland trat 1993 ein Asbestverbot in Kraft. Auch 30 Jahre später gehören Mesotheliome nach wie vor zu den häufigsten Berufskrankheiten. Diese Arbeit beleuchtet aktuelle Aspekte zu Inzidenz, Mortalität, anerkannten Berufskrankheiten und Früherkennung sowie Begutachtung.
    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)是肿瘤学的一个挑战,对晚期患者的治疗选择有限。嵌合抗原受体T细胞(CART)治疗靶向间皮素(MSLN)显示出希望,但是诸如敌对的免疫抑制肿瘤微环境(TME)之类的挑战阻碍了其疗效。这项研究探讨了在同基因PDAC模型中结合质子放射疗法(RT)与MSLN靶向CART疗法的协同潜力。质子RT显着增加肿瘤细胞中MSLN的表达,并导致CART细胞浸润到肿瘤中的显着增加。联合疗法重塑了免疫抑制TME,促进抗肿瘤M1极化巨噬细胞和减少髓源性抑制细胞(MDSC)。在侧面PDAC模型中,与单独的单独治疗相比,联合治疗表现出优异的肿瘤生长衰减和生存改善.在用图像引导质子RT处理的原位PDAC模型中,与单纯RT治疗相比,联合治疗组的肿瘤生长显著降低.Further,联合疗法在双腹肿瘤模型中诱导了远视效应,血清干扰素-γ水平升高和肿瘤外CAR-T细胞增殖增强。总之,质子RT与MSLN靶向CART疗法相结合证明可有效调节TME,增强CART细胞运输,并发挥全身抗肿瘤作用。因此,这种组合方法可以为改善不可切除PDAC的结局提供有希望的策略.
    Pancreatic ductal adenocarcinoma (PDAC) represents a challenge in oncology, with limited treatment options for advanced-stage patients. Chimeric antigen receptor T cell (CAR T) therapy targeting mesothelin (MSLN) shows promise, but challenges such as the hostile immunosuppressive tumor microenvironment (TME) hinder its efficacy. This study explores the synergistic potential of combining proton radiation therapy (RT) with MSLN-targeting CAR T therapy in a syngeneic PDAC model. Proton RT significantly increased MSLN expression in tumor cells and caused a significant increase in CAR T cell infiltration into tumors. The combination therapy reshaped the immunosuppressive TME, promoting antitumorigenic M1 polarized macrophages and reducing myeloid-derived suppressor cells (MDSC). In a flank PDAC model, the combination therapy demonstrated superior attenuation of tumor growth and improved survival compared to individual treatments alone. In an orthotopic PDAC model treated with image-guided proton RT, tumor growth was significantly reduced in the combination group compared to the RT treatment alone. Further, the combination therapy induced an abscopal effect in a dual-flank tumor model, with increased serum interferon-γ levels and enhanced proliferation of extratumoral CAR T cells. In conclusion, combining proton RT with MSLN-targeting CAR T therapy proves effective in modulating the TME, enhancing CAR T cell trafficking, and exerting systemic antitumor effects. Thus, this combinatorial approach could present a promising strategy for improving outcomes in unresectable 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
    背景:嵌合抗原受体T细胞(CAR-T)治疗已在B细胞恶性肿瘤患者中获得了显着缓解。然而,其治疗实体瘤的疗效仍然有限。这里,我们研究了使用工程长效白介素(IL)-7(rhIL-7-hyFc或NT-I7)和针对三种抗原的CAR-T细胞的联合治疗方法。磷脂酰肌醇蛋白聚糖-2(GPC2),磷脂酰肌醇蛋白聚糖-3(GPC3),和间皮素(MSLN),对抗包括肝癌在内的多种实体瘤类型,神经母细胞瘤,卵巢癌,和小鼠的胰腺癌。
    方法:靶向GPC2,GPC3和MSLN的CAR-T细胞与NT-I7联合使用以评估抗癌活性。异种移植肿瘤模型,包括肝癌原位模型,使用移植有肝细胞癌细胞系的NODscidγ小鼠建立,神经母细胞瘤,卵巢癌,还有胰腺癌.通过生物发光体内肿瘤成像和使用卡尺测量肿瘤体积来监测小鼠。对NT-I7刺激下的CAR-T细胞的免疫表型进行记忆标志物评估,排气标记,和T细胞信号分子通过流式细胞术和蛋白质印迹。
    结果:与IL-2组合相比,NT-I7的临床前评估显示实体瘤通过CD4+CAR-T的占有率增强消退,改善T细胞扩增,减少耗竭标记(程序性细胞死亡蛋白1或PD-1和淋巴细胞激活基因3或LAG-3)表达,和增加干细胞样记忆CAR-T细胞的产生。STAT5通路被证明是NT-I7信号的下游,由CAR-T细胞中IL-7受体表达的增加介导。此外,当与NT-I7联合使用时,CAR-T细胞在小鼠中提高了对低抗原密度肿瘤的功效,为具有异质性抗原特征的患者提供了一条途径。
    结论:本研究为NT-I7加CAR-T细胞联合治疗人类实体瘤提供了理论基础。
    BACKGROUND: Chimeric antigen receptor T-cell (CAR-T) therapy has achieved remarkable remission in patients with B-cell malignancies. However, its efficacy in treating solid tumors remains limited. Here, we investigated a combination therapy approach using an engineered long-acting interleukin (IL)-7 (rhIL-7-hyFc or NT-I7) and CAR-T cells targeting three antigens, glypican-2 (GPC2), glypican-3 (GPC3), and mesothelin (MSLN), against multiple solid tumor types including liver cancer, neuroblastoma, ovarian cancer, and pancreatic cancer in mice.
    METHODS: CAR-T cells targeting GPC2, GPC3, and MSLN were used in combination with NT-I7 to assess the anticancer activity. Xenograft tumor models, including the liver cancer orthotopic model, were established using NOD scid gamma mice engrafted with cell lines derived from hepatocellular carcinoma, neuroblastoma, ovarian cancer, and pancreatic cancer. The mice were monitored by bioluminescence in vivo tumor imaging and tumor volume measurement using a caliper. Immunophenotyping of CAR-T cells on NT-I7 stimulation was evaluated for memory markers, exhaust markers, and T-cell signaling molecules by flow cytometry and western blotting.
    RESULTS: Compared with the IL-2 combination, preclinical evaluation of NT-I7 exhibited regression of solid tumors via enhanced occupancy of CD4+ CAR-T, improved T-cell expansion, reduced exhaustion markers (programmed cell death protein 1 or PD-1 and lymphocyte-activation gene 3 or LAG-3) expression, and increased generation of stem cell-like memory CAR-T cells. The STAT5 pathway was demonstrated to be downstream of NT-I7 signaling, mediated by increased expression of the IL-7 receptor expression in CAR-T cells. Furthermore, CAR-T cells improved efficacy against tumors with low antigen density when combined with NT-I7 in mice, presenting an avenue for patients with heterogeneous antigenic profiles.
    CONCLUSIONS: This study provides a rationale for NT-I7 plus CAR-T cell combination therapy for solid tumors in humans.
    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
    嵌合抗原受体(CAR)-T细胞疗法是最有效的免疫疗法之一。CAR-T细胞疗法在血液系统恶性肿瘤的治疗中取得了巨大的成功。然而,由于实体恶性肿瘤的特点,如目标效应,肿瘤外毒性,免疫抑制肿瘤微环境(TME),贩运不足,CAR-T细胞治疗实体肿瘤仍处于探索阶段。间皮素(MSLN)是在各种实体恶性肿瘤细胞的表面上表达的分子,其适合作为具有用于CAR-T细胞疗法的高MSLN表达的肿瘤细胞的靶标。本文简要介绍了CAR-T细胞治疗的发展以及MSLN的结构特征,并通过总结一些临床前实验和临床试验,特别总结了MSLN靶向CAR-T细胞的结构优化策略和增强MSLN靶向CAR-T细胞抗肿瘤功效的方法。以MSLN靶向CAR-T细胞疗法为例,本文总结了研究人员在CAR-T细胞治疗实体肿瘤方面所做的努力,并通过整合现有研究成果总结出可行的治疗方案。
    Chimeric antigen receptor (CAR)-T-cell therapy is one of the most effective immunotherapies. CAR-T-cell therapy has achieved great success in the treatment of hematological malignancies. However, due to the characteristics of solid malignant tumors, such as on-target effects, off-tumor toxicity, an immunosuppressive tumor microenvironment (TME), and insufficient trafficking, CAR-T-cell therapy for solid tumors is still in the exploration stage. Mesothelin (MSLN) is a molecule expressed on the surface of various solid malignant tumor cells that is suitable as a target of tumor cells with high MSLN expression for CAR-T-cell therapy. This paper briefly described the development of CAR-T cell therapy and the structural features of MSLN, and especially summarized the strategies of structure optimization of MSLN-targeting CAR-T-cells and the enhancement methods of MSLN-targeting CAR-T cell anti-tumor efficacy by summarizing some preclinical experiment and clinical trials. When considering MSLN-targeting CAR-T-cell therapy as an example, this paper summarizes the efforts made by researchers in CAR-T-cell therapy for solid tumors and summarizes feasible treatment plans by integrating the existing research results.
    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
    上皮性卵巢癌(EOC)仍然是最致命的妇科癌症之一。细胞因子诱导的记忆样(CIML)自然杀伤(NK)细胞在临床前和早期临床试验中显示出可喜的结果。在目前的研究中,CIMLNK细胞对一组EOC细胞系表现出优异的抗肿瘤反应,激活受体的表达增加,以及与细胞周期/增殖有关的基因的上调和抑制/抑制基因的下调。用靶向间皮素(MSLN)的膜近端结构域的嵌合抗原受体(CAR)转导的CIMLNK细胞进一步改善了针对表达MSLN的EOC细胞和患者来源的异种移植肿瘤细胞的抗肿瘤反应。CIMLNK细胞的CAR武装实质上减少了患者来源的腹水液中的功能障碍,与代谢改变和补品信号传导相关的转录组变化作为潜在机制。最后,MSLN-CARCIMLNK细胞的过继转移证明了对肿瘤生长的显着抑制作用,并阻止了异种移植小鼠的转移扩散,支持他们作为EOC有效治疗策略的潜力。
    Epithelial ovarian cancer (EOC) remains one of the most lethal gynecological cancers. Cytokine-induced memory-like (CIML) natural killer (NK) cells have shown promising results in preclinical and early-phase clinical trials. In the current study, CIML NK cells demonstrated superior antitumor responses against a panel of EOC cell lines, increased expression of activation receptors, and up-regulation of genes involved in cell cycle/proliferation and down-regulation of inhibitory/suppressive genes. CIML NK cells transduced with a chimeric antigen receptor (CAR) targeting the membrane-proximal domain of mesothelin (MSLN) further improved the antitumor responses against MSLN-expressing EOC cells and patient-derived xenograft tumor cells. CAR arming of the CIML NK cells subtanstially reduced their dysfunction in patient-derived ascites fluid with transcriptomic changes related to altered metabolism and tonic signaling as potential mechanisms. Lastly, the adoptive transfer of MSLN-CAR CIML NK cells demonstrated remarkable inhibition of tumor growth and prevented metastatic spread in xenograft mice, supporting their potential as an effective therapeutic strategy in EOC.
    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
    除了它们的免疫抑制作用,在过继治疗之前的细胞抑制剂调节,例如嵌合抗原受体(CAR)T细胞可能在肿瘤微环境的减积和重塑中起作用。我们在体外研究了曲硫丹和氟达拉滨对表达间皮素(MSLN)的卵巢癌细胞的杀伤功效和影响,以及对MSLN靶向CART细胞的影响。曲硫丹和氟达拉滨对SKOV3和OVCAR4细胞具有协同杀伤作用。当SKOV3细胞表达MSLN和OVCAR4细胞在缺氧条件下测试时,对曲硫丹和氟达拉滨组合的敏感性增加,而曲硫丹或氟达拉滨暴露后,SKOV3和OVCAR4细胞的MSLN细胞表面表达没有改变。暴露于曲硫丹或氟达拉滨(10µM)均不影响MSLN-CART细胞脱颗粒,用MSLN+OVCAR3细胞攻击后产生细胞因子,也不诱导线粒体缺陷。曲硫丹和氟达拉滨的组合降低了常氧下的MSLN-CART细胞抗肿瘤杀伤,但不降低缺氧。总之,曲硫丹和氟达拉滨杀死MSLN+卵巢癌细胞而不改变MSLN-CAR-T细胞的功能(在低细胞抑制剂浓度下),即使在缺氧条件下,我们的数据支持在MSLN-CAR-T细胞治疗前使用曲硫丹和氟达拉滨作为调理药物.
    In addition to their immunosuppressive effect, cytostatics conditioning prior to adoptive therapy such as chimeric antigen receptor (CAR) T cells may play a role in debulking and remodeling the tumor microenvironment. We investigated in vitro the killing efficacy and impact of treosulfan and fludarabine on ovarian cancer cells expressing mesothelin (MSLN) and effect on MSLN-targeting CAR T cells. Treosulfan and fludarabine had a synergetic effect on killing of SKOV3 and OVCAR4 cells. Sensitivity to the combination of treosulfan and fludarabine was increased when SKOV3 cells expressed MSLN and when OVCAR4 cells were tested in hypoxia, while MSLN cells surface expression by SKOV3 and OVCAR4 cells was not altered after treosulfan or fludarabine exposure. Exposure to treosulfan or fludarabine (10 µM) neither impacted MSLN-CAR T cells degranulation, cytokines production upon challenge with MSLN + OVCAR3 cells, nor induced mitochondrial defects. Combination of treosulfan and fludarabine decreased MSLN-CAR T cells anti-tumor killing in normoxia but not hypoxia. In conclusion, treosulfan and fludarabine killed MSLN + ovarian cancer cells without altering MSLN-CAR T cells functions (at low cytostatics concentration) even in hypoxic conditions, and our data support the use of treosulfan and fludarabine as conditioning drugs prior to MSLN-CAR T cell 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: Letter
    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
    Fangchinoline(FA)是一种生物碱,来源于中药方济。大量研究表明,FA对各种癌细胞具有毒性作用,但是对它对生殖细胞的毒性作用知之甚少,尤其是卵母细胞。在这项研究中,我们研究了FA对小鼠卵母细胞成熟的影响及其潜在机制。我们的结果表明,FA不会影响减数分裂的恢复,但会抑制第一极体的挤压。这种抑制不是由于细胞器水平的异常,比如染色体和线粒体,DNA损伤和活性氧的检测证明了这一点。进一步的研究表明,FA在中期I阶段阻止了卵母细胞,这种逮捕不是由动粒-微管连接异常或纺锤体组装检查点激活引起的。相反,FA抑制后期促进复合物(APC/C)的活性,如CCNB1变性的抑制所证明的。APC/C的活性降低可能是由于CDC25B活性降低,如CDC25B(Ser323)的高磷酸化水平所示。这可以进一步增强成熟促进因子(MPF)活性,在减数分裂中起关键作用。总之,我们的研究提示,FA引起的中期Ⅰ期阻滞可能是由于MPF和APC/C活性异常所致.
    Fangchinoline (FA) is an alkaloid derived from the traditional Chinese medicine Fangji. Numerous studies have shown that FA has a toxic effect on various cancer cells, but little is known about its toxic effects on germ cells, especially oocytes. In this study, we investigated the effects of FA on mouse oocyte maturation and its potential mechanisms. Our results showed that FA did not affect meiosis resumption but inhibited the first polar body extrusion. This inhibition is not due to abnormalities at the organelle level, such as chromosomes and mitochondrial, which was proved by detection of DNA damage and reactive oxygen species. Further studies revealed that FA arrested the oocyte at the metaphase I stage, and this arrest was not caused by abnormal kinetochore-microtubule attachment or spindle assembly checkpoint activation. Instead, FA inhibits the activity of anaphase-promoting complexes (APC/C), as evidenced by the inhibition of CCNB1 degeneration. The decreased activity of APC/C may be due to a reduction in CDC25B activity as indicated by the high phosphorylation level of CDC25B (Ser323). This may further enhance Maturation-Promoting Factor (MPF) activity, which plays a critical role in meiosis. In conclusion, our study suggests that the metaphase I arrest caused by FA may be due to abnormalities in MPF and APC/C activity.
    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
    T细胞接合双特异性抗体(TCB)最近在癌症治疗中变得重要。在这项研究中,我们开发了MSLN490,一种新型的TCB,旨在靶向间皮素(MSLN),糖基磷脂酰肌醇(GPI)连接的糖蛋白在各种癌症中高表达,并评估了其对实体瘤的疗效。使用CDR步移和噬菌体展示技术来改善亲本抗体M912的亲和力,产生对MSLN具有不同亲和力的抗体池。从这个游泳池里,组装各种双特异性抗体(BsAb)。值得注意的是,具有其IgG-[L]-scFv结构的MSLN490对表达MSLN的肿瘤显示出显著的抗肿瘤活性(在HT-29-hMSLN细胞中EC50:0.16pM)。此外,即使在非膜锚定的MSLN(可溶性MSLN)存在下,MSLN490仍然有效。此外,MSLN490与Atezolizumab或TAA×CD28BsAb联合使用时,其抗肿瘤活性增强.值得注意的是,在MSLN490和紫杉醇之间观察到协同作用,由于紫杉醇破坏了实体瘤中的免疫抑制微环境,增强免疫细胞浸润,提高抗肿瘤疗效。总的来说,MSLN490具有强大的抗肿瘤活性,抗可溶性MSLN干扰,与其他疗法结合使用时,增强了抗肿瘤作用,为各种实体瘤的治疗提供了一个有希望的未来。本研究为进一步探索MSLN490的临床潜力奠定了坚实的基础。
    T cell engaging bispecific antibodies (TCBs) have recently become significant in cancer treatment. In this study we developed MSLN490, a novel TCB designed to target mesothelin (MSLN), a glycosylphosphatidylinositol (GPI)-linked glycoprotein highly expressed in various cancers, and evaluated its efficacy against solid tumors. CDR walking and phage display techniques were used to improve affinity of the parental antibody M912, resulting in a pool of antibodies with different affinities to MSLN. From this pool, various bispecific antibodies (BsAbs) were assembled. Notably, MSLN490 with its IgG-[L]-scFv structure displayed remarkable anti-tumor activity against MSLN-expressing tumors (EC50: 0.16 pM in HT-29-hMSLN cells). Furthermore, MSLN490 remained effective even in the presence of non-membrane-anchored MSLN (soluble MSLN). Moreover, the anti-tumor activity of MSLN490 was enhanced when combined with either Atezolizumab or TAA × CD28 BsAbs. Notably, a synergistic effect was observed between MSLN490 and paclitaxel, as paclitaxel disrupted the immunosuppressive microenvironment within solid tumors, enhancing immune cells infiltration and improved anti-tumor efficacy. Overall, MSLN490 exhibits robust anti-tumor activity, resilience to soluble MSLN interference, and enhanced anti-tumor effects when combined with other therapies, offering a promising future for the treatment of a variety of solid tumors. This study provides a strong foundation for further exploration of MSLN490\'s clinical potential.
    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