oncolytic virotherapy

溶瘤病毒疗法
  • 文章类型: Journal Article
    肝细胞癌是一种难治性肿瘤,预后差,死亡率高。目前正在研究许多溶瘤病毒用于治疗肝细胞癌。根据以前的研究,我们构建了携带GM-CSF的辛德毕斯病毒,命名为SINV-GM-CSF,其在病毒载体的nsp1蛋白的氨基酸285处含有突变(G到S)。在细胞水平和体内验证了这种突变载体用于肝癌治疗的潜力,分别,并描述了治疗后肿瘤微环境的变化。结果表明,辛德毕斯病毒能有效感染肝癌细胞系并诱导细胞死亡。此外,GM-CSF的加入增强了辛德毕斯病毒的肿瘤杀伤作用,并增加了治疗期间肿瘤内微环境中免疫细胞的数量.特别是,在肝细胞癌的小鼠肿瘤模型中,SINV-GM-CSF能够通过调节M1型巨噬细胞(其具有肿瘤抗性能力)的升高和M2型巨噬细胞(其具有肿瘤促进能力)的减少而有效地杀死肿瘤。总的来说,SINV-GM-CSF是一种有吸引力的载体平台,具有作为安全有效的溶瘤病毒的临床潜力。
    Hepatocellular carcinoma is a refractory tumor with poor prognosis and high mortality. Many oncolytic viruses are currently being investigated for the treatment of hepatocellular carcinoma. Based on previous studies, we constructed a recombinant GM-CSF-carrying Sindbis virus, named SINV-GM-CSF, which contains a mutation (G to S) at amino acid 285 in the nsp1 protein of the viral vector. The potential of this mutated vector for liver cancer therapy was verified at the cellular level and in vivo, respectively, and the changes in the tumor microenvironment after treatment were also described. The results showed that the Sindbis virus could effectively infect hepatocellular carcinoma cell lines and induce cell death. Furthermore, the addition of GM-CSF enhanced the tumor-killing effect of the Sindbis virus and increased the number of immune cells in the intra-tumor microenvironment during the treatment. In particular, SINV-GM-CSF was able to efficiently kill tumors in a mouse tumor model of hepatocellular carcinoma by regulating the elevation of M1-type macrophages (which have a tumor-resistant ability) and the decrease in M2-type macrophages (which have a tumor-promoting capacity). Overall, SINV-GM-CSF is an attractive vector platform with clinical potential for use as a safe and effective oncolytic virus.
    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
    根据世界卫生组织的数据,癌症仍然是致命疾病的最前沿。全球观察到癌症发病率和死亡率呈上升趋势,强调应继续努力开发检测和治疗方法。诊断路径通常从学习患者的病史开始;随后是基本的血液检查和成像测试,以指示癌症可能位于何处,以安排穿刺活检。迅速开始诊断至关重要,因为延迟的癌症检测需要更高的治疗和住院成本。因此,需要新的癌症检测方法,如液体活检,弹性成像,合成生物传感器,荧光成像,和反射共聚焦显微镜。常规治疗方法,虽然在临床实践中仍然很常见,存在许多局限性,并且不能令人满意。如今,临床研究的动态发展和更精确和有效的方法的发展,如溶瘤病毒疗法,基于外泌体的治疗,纳米技术,树突状细胞,嵌合抗原受体,免疫检查点抑制剂,基于天然产品的治疗,肿瘤治疗领域,和光动力疗法。本文比较了有关常规和现代癌症检测和治疗方法的可用数据,以促进对这一迅速发展的领域及其未来方向的理解。事实证明,现代方法并非没有缺点;仍然需要开发新的检测策略和治疗方法来提高灵敏度,特异性,安全,和功效。然而,已经采取了适当的路线,食品和药物管理局批准了一些现代方法。
    In accordance with the World Health Organization data, cancer remains at the forefront of fatal diseases. An upward trend in cancer incidence and mortality has been observed globally, emphasizing that efforts in developing detection and treatment methods should continue. The diagnostic path typically begins with learning the medical history of a patient; this is followed by basic blood tests and imaging tests to indicate where cancer may be located to schedule a needle biopsy. Prompt initiation of diagnosis is crucial since delayed cancer detection entails higher costs of treatment and hospitalization. Thus, there is a need for novel cancer detection methods such as liquid biopsy, elastography, synthetic biosensors, fluorescence imaging, and reflectance confocal microscopy. Conventional therapeutic methods, although still common in clinical practice, pose many limitations and are unsatisfactory. Nowadays, there is a dynamic advancement of clinical research and the development of more precise and effective methods such as oncolytic virotherapy, exosome-based therapy, nanotechnology, dendritic cells, chimeric antigen receptors, immune checkpoint inhibitors, natural product-based therapy, tumor-treating fields, and photodynamic therapy. The present paper compares available data on conventional and modern methods of cancer detection and therapy to facilitate an understanding of this rapidly advancing field and its future directions. As evidenced, modern methods are not without drawbacks; there is still a need to develop new detection strategies and therapeutic approaches to improve sensitivity, specificity, safety, and efficacy. Nevertheless, an appropriate route has been taken, as confirmed by the approval of some modern methods by the Food and Drug Administration.
    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
    前列腺癌是男性中普遍存在的癌症,和传统的治疗选择往往是有限的。细胞毒性化疗,尽管它有缺点,仍然是中流砥柱。我们提出了一种针对溶瘤麻疹病毒(OMV)和长春新碱(VC)的纳米级递送单位的靶向共递送方法,以增强治疗功效。HA包被的OMV+VC负载的TC纳米制剂被设计用于前列腺癌中的靶向溶瘤活性。前列腺癌细胞系中的CD44表达分析表明在PC3细胞中显著高表达。使用专家设计(DOE)进行纳米配方的优化,和HA包被的OMV+VC负载的TC纳米制剂的制备和表征被详细描述,显示平均粒度397.2±0.01nm和多分散指数0.122,ζ电位19.7±0.01mV。结果表明,成功的包封效率为2.4×106TCID50/Ml,并且从纳米制剂中持续释放OMV和VC长达72小时。分析显示,PC3细胞在10±0.71%的细胞活力下具有有效的抗癌活性,而在HPrEC中为73±0.66%,在剂量和时间依赖性方式为90µg/ml时具有显着的形态学变化。共制剂在PBS中50µgPI/ml时显示阳性细胞死亡49.5±0.02%,在G2/M期细胞周期停滞54.3%,8.1%G0/G1和5.7%在S阶段,具有50µg/ml的显着线粒体膜电位(MMP),通过流式细胞术(FACS)评估。表面整合配体方法增强溶瘤病毒和化疗药物的靶向递送,提出了前列腺癌治疗的潜在替代方案,并表明在纳米制剂中共同施用VC和OMV可以改善治疗结果,同时减少化疗药物剂量。
    Prostate cancer is a prevalent carcinoma among males, and conventional treatment options are often limited. Cytotoxic chemotherapy, despite its drawbacks, remains a mainstay. We propose a targeted co-delivery approach using nanoscale delivery units for Oncolytic measles virus (OMV) and vincristine (VC) to enhance treatment efficacy. The HA-coated OMV + VC-loaded TCs nanoformulation is designed for targeted oncolytic activity in prostate cancer. The CD44 expression analysis in prostate cancer cell lines indicates a significantly high expression in PC3 cells. The optimization of nanoformulations using Design of Expert (DOE) is performed, and the preparation and characterization of HA-coated OMV + VC-loaded TCs nanoformulations are detailed showing average particle size 397.2 ± 0.01 nm and polydispersity index 0.122 with zeta potential 19.7 + 0.01 mV. Results demonstrate successful encapsulation efficiency with 2.4 × 106 TCID50/Ml and sustained release of OMV and VC from the nanoformulation for up to 72 h. In vitro, assays reveal potent anticancer activity at 10 ± 0.71% cell viability in PC3 cells compared to 73 ± 0.66% in HPrEC and significant morphological changes at 90 µg/ml in dose and time-dependent manner. The co-formulation showed positive cell death 49.5 ± 0.02% at 50 µg PI/ml in PBS and 54.3% cell cycle arrest at the G2/M phase, 8.1% G0/G1 and 5.7% at S phase, with significant mitochondrial membrane potential (MMP) at 50 µg/ml, as assessed by flow cytometry (FACS). The surface-integrating ligand approach enhances the targeted delivery of the oncolytic virus and chemotherapeutic drug, presenting a potential alternative for prostate cancer treatment and suggested that co-administering VC and OMV in a nanoformulation could improve therapeutic outcomes while reducing chemotherapeutic drug doses.
    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
    最近的研究表明,溶瘤病毒与CAR-T细胞联合治疗已显示出优异的抗肿瘤效果,代表一种有希望的方法。尽管如此,肿瘤内注射的局部给药方法对治疗难以触及的转移性肿瘤或远端肿瘤提出了挑战.为了解决这个障碍,我们使用HSV-1感染的CAR-T细胞,将HSV全身递送到实体瘤中。在用HSV加载CAR-T细胞三天后,其生物学功能保持完整。在免疫受损和免疫活性GBM原位小鼠模型中,B7-H3CAR-T细胞有效地将HSV递送至肿瘤病变,导致T细胞浸润增强,并显着延长小鼠的生存期。我们还采用了双侧皮下肿瘤模型,观察到接受肿瘤内病毒注射的组在注射侧肿瘤体积显着减少,而接受静脉输注携带HSV的CAR-T细胞的组显示两侧肿瘤生长受到抑制。因此,CAR-THSV细胞在HSV向远处肿瘤的全身递送中提供显著优势。总之,我们的发现强调了CAR-T细胞作为HSV携带者的潜力,针对远处肿瘤的溶瘤病毒疗法具有显著优势。
    Recent studies have indicated that combining oncolytic viruses with CAR-T cells in therapy has shown superior anti-tumor effects, representing a promising approach. Nonetheless, the localized delivery method of intratumoral injection poses challenges for treating metastatic tumors or distal tumors that are difficult to reach. To address this obstacle, we employed HSV-1-infected CAR-T cells, which systemically delivery HSV into solid tumors. The biological function of CAR-T cells remained intact after loading them with HSV for a period of three days. In both immunocompromised and immunocompetent GBM orthotopic mouse models, B7-H3 CAR-T cells effectively delivered HSV to tumor lesions, resulting in enhanced T-cell infiltration and significantly prolonged survival in mice. We also employed a bilateral subcutaneous tumor model and observed that the group receiving intratumoral virus injection exhibited a significant reduction in tumor volume on the injected side, while the group receiving intravenous infusion of CAR-T cells carrying HSV displayed suppressed tumor growth on both sides. Hence, CAR-THSV cells offer notable advantages in the systemic delivery of HSV to distant tumors. In conclusion, our findings emphasize the potential of CAR-T cells as carriers for HSV, presenting significant advantages for oncolytic virotherapy targeting distant tumors.
    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:胰腺导管腺癌(PDAC)是一种未满足医疗需求的肿瘤实体。为了评估溶瘤病毒疗法(OVT)对PDAC的治疗潜力,目前在临床试验中研究了不同的溶瘤病毒(OVs)。然而,这些不同OVs在抗PDAC疗效和预测治疗反应的生物标志物方面缺乏系统比较.
    方法:我们筛选了14种患者来源的PDAC培养物,这些培养物反映了PDAC对5种临床相关OVs的敏感性,即血清型5腺病毒Ad5-hTERT,疱疹病毒T-VEC,麻疹疫苗株MV-NIS,呼肠孤病毒jin-3和原病毒H-1PV。活细胞分析,病毒基因组/基因表达的定量,进行细胞活力以及细胞毒性测定和病毒后代的滴定。转录组分析用于鉴定对OV治疗的反应的潜在预测性生物标志物。
    结果:患者来源的PDAC培养物显示出对OV治疗的个体反应模式。十四个培养物中有十二个对至少一个OV有反应,在所有文化中,没有单一的OV被证明是优越还是低劣。检索不同病毒的已知宿主因子作为潜在生物标志物。与经典分子亚型相比,发现PDAC的准间充质或基底样亚型对H-1PV更敏感,jin-3和T-VEC。一般来说,病毒进入受体的表达与OV治疗的敏感性无关,有一个例外:半乳糖凝集素-1(LGALS1)的表达,参与H-1PV进入的一个因素,与H-1PV诱导的细胞杀伤呈正相关。相反,细胞途径控制免疫学,代谢和增殖信号似乎决定了结局.例如,干扰素刺激基因(ISGs)的高基线表达与溶瘤麻疹病毒的相对抗性相关,而低环状GMP-AMP合酶(cGAS)表达与异常反应相关。MV-NIS与cGAS抑制剂的组合治疗可改善几种PDAC培养物中的肿瘤细胞杀伤,并且发现过表达cGAS的细胞对MV溶瘤的敏感性较低。
    结论:考虑到PDAC的异质性和OVs等生物疗法的复杂性,没有单一的生物标志物可以解释反应模式的频谱.为了选择特定的OV,PDAC分子亚型,应考虑ISG表达以及不同信号传导和代谢途径的激活。组合疗法可以克服特定星座中的抗性。总的来说,溶瘤病毒疗法是PDAC的可行治疗选择,这需要进一步发展。这项研究强调了OVT个性化治疗的必要性。通过提供所有主要数据,这项研究为正在进行的发展提供了丰富的资源和指导。
    背景:德国国家科学基金会(DeutscheForschungsgemeinschaft,DFG),德国癌症援助组织(DeutscheKrebshilfe),德国国家学术奖学金基金会(StudienstiftungdesdeutschenVolkes),胰腺癌基金会的生存。
    BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is a tumour entity with unmet medical need. To assess the therapeutic potential of oncolytic virotherapy (OVT) against PDAC, different oncolytic viruses (OVs) are currently investigated in clinical trials. However, systematic comparisons of these different OVs in terms of efficacy against PDAC and biomarkers predicting therapeutic response are lacking.
    METHODS: We screened fourteen patient-derived PDAC cultures which reflect the intra- and intertumoural heterogeneity of PDAC for their sensitivity to five clinically relevant OVs, namely serotype 5 adenovirus Ad5-hTERT, herpes virus T-VEC, measles vaccine strain MV-NIS, reovirus jin-3, and protoparvovirus H-1PV. Live cell analysis, quantification of viral genome/gene expression, cell viability as well as cytotoxicity assays and titration of viral progeny were conducted. Transcriptome profiling was employed to identify potential predictive biomarkers for response to OV treatment.
    RESULTS: Patient-derived PDAC cultures showed individual response patterns to OV treatment. Twelve of fourteen cultures were responsive to at least one OV, with no single OV proving superior or inferior across all cultures. Known host factors for distinct viruses were retrieved as potential biomarkers. Compared to the classical molecular subtype, the quasi-mesenchymal or basal-like subtype of PDAC was found to be more sensitive to H-1PV, jin-3, and T-VEC. Generally, expression of viral entry receptors did not correlate with sensitivity to OV treatment, with one exception: Expression of Galectin-1 (LGALS1), a factor involved in H-1PV entry, positively correlated with H-1PV induced cell killing. Rather, cellular pathways controlling immunological, metabolic and proliferative signaling appeared to determine outcome. For instance, high baseline expression of interferon-stimulated genes (ISGs) correlated with relative resistance to oncolytic measles virus, whereas low cyclic GMP-AMP synthase (cGAS) expression was associated with exceptional response. Combination treatment of MV-NIS with a cGAS inhibitor improved tumour cell killing in several PDAC cultures and cells overexpressing cGAS were found to be less sensitive to MV oncolysis.
    CONCLUSIONS: Considering the heterogeneity of PDAC and the complexity of biological therapies such as OVs, no single biomarker can explain the spectrum of response patterns. For selection of a particular OV, PDAC molecular subtype, ISG expression as well as activation of distinct signaling and metabolic pathways should be considered. Combination therapies can overcome resistance in specific constellations. Overall, oncolytic virotherapy is a viable treatment option for PDAC, which warrants further development. This study highlights the need for personalised treatment in OVT. By providing all primary data, this study provides a rich source and guidance for ongoing developments.
    BACKGROUND: German National Science Foundation (Deutsche Forschungsgemeinschaft, DFG), German Cancer Aid (Deutsche Krebshilfe), German National Academic Scholarship Foundation (Studienstiftung des deutschen Volkes), Survival with Pancreatic Cancer Foundation.
    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
    尽管患者在多种肿瘤中受益于免疫检查点抑制(ICI)治疗,耐药性可能来自免疫抑制肿瘤微环境(TME),这是特别真实的肝细胞癌(HCC)。由于溶瘤病毒(OV)可以产生高度免疫浸润,炎性TME,OV可能会通过招募来恢复ICI的响应能力,启动,和激活抗肿瘤T细胞。在这里,我们发现相反,溶瘤性水疱性口炎病毒,表达干扰素β(VSV-IFNβ),在部分抗PD-L1反应性HCC模型中拮抗抗PD-L1治疗的效果。通过飞行时间的细胞测定法显示,VSV-IFNβ扩增显性抗病毒效应CD8T细胞,伴随着抗肿瘤T细胞群的相对消失,它们是抗PD-L1的靶标。然而,通过在VSV内表达一系列HCC肿瘤抗原,OV和抗PD-L1联合治疗获益可以恢复。我们的数据通过显示显性抗病毒T细胞应答可以抑制亚显性抗肿瘤T细胞应答,为使用高免疫原性病毒作为肿瘤特异性免疫治疗剂提供了警示信息。然而,通过在病毒中编码肿瘤抗原,溶瘤病毒疗法可以产生抗肿瘤T细胞群体,免疫检查点阻断可以有效地发挥作用。
    Although patients benefit from immune checkpoint inhibition (ICI) therapy in a broad variety of tumors, resistance may arise from immune suppressive tumor microenvironments (TME), which is particularly true of hepatocellular carcinoma (HCC). Since oncolytic viruses (OV) can generate a highly immune-infiltrated, inflammatory TME, OVs could potentially restore ICI responsiveness via recruitment, priming, and activation of anti-tumor T cells. Here we find that on the contrary, an oncolytic vesicular stomatitis virus, expressing interferon-ß (VSV-IFNß), antagonizes the effect of anti-PD-L1 therapy in a partially anti-PD-L1-responsive model of HCC. Cytometry by Time of Flight shows that VSV-IFNß expands dominant anti-viral effector CD8 T cells with concomitant relative disappearance of anti-tumor T cell populations, which are the target of anti-PD-L1. However, by expressing a range of HCC tumor antigens within VSV, combination OV and anti-PD-L1 therapeutic benefit could be restored. Our data provide a cautionary message for the use of highly immunogenic viruses as tumor-specific immune-therapeutics by showing that dominant anti-viral T cell responses can inhibit sub-dominant anti-tumor T cell responses. However, through encoding tumor antigens within the virus, oncolytic virotherapy can generate anti-tumor T cell populations upon which immune checkpoint blockade can effectively work.
    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
    溶瘤病毒疗法,使用水泡性口炎病毒(VSVΔ51)和单纯疱疹病毒-1(HSV-1)等病毒选择性攻击癌细胞,面临挑战,如干扰素(IFN)反应介导的细胞抗性。富马酸二甲酯(DMF)用于治疗多发性硬化症和牛皮癣,并因其抗癌特性而得到认可,并已显示可增强VSVΔ51和HSV-1溶瘤活性。富马酸泰匹胺(TPF)是目前正在进行临床试验的DMF类似物,用于治疗中度至重度斑块状牛皮癣。这项研究的目的是评估TPF增强溶瘤病毒有效性的潜力。体外,TPF治疗使786-0癌细胞更容易感染VSVΔ51,导致病毒复制增加。在增加病毒感染和增加这些抗性癌细胞和其他测试的癌细胞系的杀伤方面,它优于DMF。离体研究表明TPF选择性增强癌细胞中的溶瘤病毒感染而不影响健康组织。在胰腺和卵巢肿瘤样品中的有效性显著高。我们的研究进一步表明,TPF可以通过类似于DMF的机制下调IFN途径,使耐药癌细胞更容易受到病毒感染。此外,评估了TPF对基因治疗的影响,揭示了其增强慢病毒等载体转导效率的能力,腺病毒5型和腺相关病毒2型跨各种细胞系。该数据强调了TPF不仅在溶瘤病毒治疗中而且在基因治疗的更广泛的应用中的潜在作用。总的来说,这些发现将TPF定位为溶瘤病毒疗法中的有前途的药物,保证进一步探索其治疗潜力。
    Oncolytic virotherapy, using viruses such as vesicular stomatitis virus (VSVΔ51) and Herpes Simplex Virus-1 (HSV-1) to selectively attack cancer cells, faces challenges such as cellular resistance mediated by the interferon (IFN) response. Dimethyl fumarate (DMF) is used in the treatment of multiple sclerosis and psoriasis and is recognized for its anti-cancer properties and has been shown to enhance both VSVΔ51 and HSV-1 oncolytic activity. Tepilamide fumarate (TPF) is a DMF analog currently undergoing clinical trials for the treatment of moderate-to-severe plaque psoriasis. The aim of this study was to evaluate the potential of TPF in enhancing the effectiveness of oncolytic viruses. In vitro, TPF treatment rendered 786-0 carcinoma cells more susceptible to VSVΔ51 infection, leading to increased viral replication. It outperformed DMF in both increasing viral infection and increasing the killing of these resistant cancer cells and other cancer cell lines tested. Ex vivo studies demonstrated TPF\'s selective boosting of oncolytic virus infection in cancer cells without affecting healthy tissues. Effectiveness was notably high in pancreatic and ovarian tumor samples. Our study further indicates that TPF can downregulate the IFN pathway through a similar mechanism to DMF, making resistant cancer cells more vulnerable to viral infection. Furthermore, TPF\'s impact on gene therapy was assessed, revealing its ability to enhance the transduction efficiency of vectors such as lentivirus, adenovirus type 5, and adeno-associated virus type 2 across various cell lines. This data underscore TPF\'s potential role in not only oncolytic virotherapy but also in the broader application of gene therapy. Collectively, these findings position TPF as a promising agent in oncolytic virotherapy, warranting further exploration of its therapeutic potential.
    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
    新城疫病毒(NDV)是一种禽类病原体,具有未分段的负链RNA基因组,属于副粘病毒科。虽然主要在鸟类中致病,NDV对人类健康没有威胁,使其成为各种生物医学应用的安全候选物。广泛的研究强调了NDV作为疫苗开发和基因治疗载体的潜力,由于其转录模块化,低重组率,复制过程中缺乏DNA阶段。此外,NDV表现出溶瘤能力,有效引发抗肿瘤免疫反应,从而将其定位为癌症治疗的有前途的治疗剂。本文综合评述了新城疫病毒的生物学特性,阐明了其溶瘤特性的分子机制,并讨论了其在疫苗载体开发和肿瘤治疗领域的应用。
    Newcastle disease virus (NDV) is an avian pathogen with an unsegmented negative-strand RNA genome that belongs to the Paramyxoviridae family. While primarily pathogenic in birds, NDV presents no threat to human health, rendering it a safe candidate for various biomedical applications. Extensive research has highlighted the potential of NDV as a vector for vaccine development and gene therapy, owing to its transcriptional modularity, low recombination rate, and lack of a DNA phase during replication. Furthermore, NDV exhibits oncolytic capabilities, efficiently eliciting antitumor immune responses, thereby positioning it as a promising therapeutic agent for cancer treatment. This article comprehensively reviews the biological characteristics of NDV, elucidates the molecular mechanisms underlying its oncolytic properties, and discusses its applications in the fields of vaccine vector development and tumor 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
    胶质母细胞瘤是最常见和致命的中枢神经系统恶性肿瘤,其进展后的中位生存期仅为6-9个月。胶质母细胞瘤复发的主要生化机制包括异常的分子途径,诱发复发的肿瘤微环境,和表观遗传修饰。当代护理标准(手术,辐射,化疗,和肿瘤治疗领域)有助于控制原发性肿瘤,但很少防止复发。手术等细胞减灭术治疗对复发性胶质母细胞瘤有益处;然而,它的使用仍然存在争议。复发性胶质母细胞瘤正在出现几种创新的治疗方法,包括检查点抑制剂,嵌合抗原受体T细胞疗法,溶瘤病毒疗法,纳米颗粒递送,激光间质热疗,和光动力疗法。这篇综述旨在为读者提供以下概述:(1)复发分子基础的最新发现;(2)手术在治疗复发中的作用;(3)复发性胶质母细胞瘤的新治疗范例。
    Glioblastoma is the most common and lethal central nervous system malignancy with a median survival after progression of only 6-9 months. Major biochemical mechanisms implicated in glioblastoma recurrence include aberrant molecular pathways, a recurrence-inducing tumor microenvironment, and epigenetic modifications. Contemporary standard-of-care (surgery, radiation, chemotherapy, and tumor treating fields) helps to control the primary tumor but rarely prevents relapse. Cytoreductive treatment such as surgery has shown benefits in recurrent glioblastoma; however, its use remains controversial. Several innovative treatments are emerging for recurrent glioblastoma, including checkpoint inhibitors, chimeric antigen receptor T cell therapy, oncolytic virotherapy, nanoparticle delivery, laser interstitial thermal therapy, and photodynamic therapy. This review seeks to provide readers with an overview of (1) recent discoveries in the molecular basis of recurrence; (2) the role of surgery in treating recurrence; and (3) novel treatment paradigms emerging for recurrent glioblastoma.
    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
    CAR-T细胞疗法为延长癌症缓解提供了一种有希望的方法,特别是在血癌的情况下。然而,其在实体瘤治疗中的应用仍面临诸多局限。这篇综述文章全面概述了与CAR-T细胞治疗实体肿瘤相关的挑战和策略。专注于妇科癌症。这项研究讨论了CAR-T疗法用于实体瘤治疗的局限性,比如T细胞衰竭,基质屏障,和抗原脱落。此外,它提出了增加实体肿瘤中CAR-T功效的可能方法,包括检查点抑制剂和化疗的联合治疗,以及将CAR-T与溶瘤病毒疗法相结合的新方法。鉴于缺乏对CAR-T联合疗法治疗妇科癌症的全面研究,这篇综述旨在为实体瘤联合治疗的现状提供见解,并强调这种方法在妇科中的潜力。
    CAR-T cell therapy offers a promising way for prolonged cancer remission, specifically in the case of blood cancers. However, its application in the treatment of solid tumors still faces many limitations. This review paper provides a comprehensive overview of the challenges and strategies associated with CAR-T cell therapy for solid tumors, with a focus on gynecological cancer. This study discusses the limitations of CAR-T therapy for solid tumor treatment, such as T cell exhaustion, stromal barrier, and antigen shedding. Additionally, it addresses possible approaches to increase CAR-T efficacy in solid tumors, including combination therapies with checkpoint inhibitors and chemotherapy, as well as the novel approach of combining CAR-T with oncolytic virotherapy. Given the lack of comprehensive research on CAR-T combination therapies for treating gynecological cancers, this review aims to provide insights into the current landscape of combination therapies for solid tumors and highlight the potential of such an approach in gynecology.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号