VSVΔ51

VSVΔ 51
  • 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
    建立的HER2+癌症小鼠模型是基于啮齿动物Neu/Erbb2同源物的过表达,与人类HER2(huHER2)靶向治疗不相容。此外,免疫缺陷异种移植或转基因模型的使用排除了对天然抗肿瘤免疫应答的评估.这些障碍对我们理解huHER2靶向免疫疗法背后的免疫机制是一个挑战。
    为了评估我们的huHER2靶向组合策略的免疫影响,我们建立了huHER2+乳腺癌的同基因小鼠模型,使用截短形式的huHER2,HER2。在验证此模型后,接下来,我们采用我们的免疫治疗策略治疗荷瘤患者:溶瘤性水泡性口炎病毒(VSVΔ51)和临床批准的抗体-药物偶联物靶向huHER2曲妥珠单抗emtansine(T-DM1).我们通过肿瘤控制来评估疗效,生存,和免疫分析。
    当在鼠乳腺癌4T1.2细胞中表达时,产生的截短的HER2T构建体在野生型BALB/c小鼠中是非免疫原性的。与对照组相比,用VSVΔ51+T-DM1治疗4T1.2-HER2T肿瘤产生了强大的疗效,和广泛的免疫记忆。抗肿瘤免疫的询问显示CD4+T细胞浸润肿瘤,和B的激活,NK,和树突状细胞反应,以及肿瘤反应性血清IgG。
    4T1.2-HER2T模型用于评估我们的复杂药物病毒治疗策略后的抗肿瘤免疫应答。这些数据证明了同基因HER2T模型在体内免疫活性设置中用于评估huHER2靶向治疗的实用性。我们进一步证明,HER2T可以在多个其他同系肿瘤模型中实施,包括但不限于结直肠和卵巢模型。这些数据还表明,HER2T平台可用于评估一系列表面HER2T靶向方法。比如CAR-T,T细胞衔接者,抗体,甚至重新定位溶瘤病毒。
    Established mouse models of HER2+ cancer are based on the over-expression of rodent Neu/Erbb2 homologues, which are incompatible with human HER2 (huHER2) targeted therapeutics. Additionally, the use of immune-deficient xenograft or transgenic models precludes assessment of native anti-tumour immune responses. These hurdles have been a challenge for our understanding of the immune mechanisms behind huHER2-targeting immunotherapies.
    To assess the immune impacts of our huHER2-targeted combination strategy, we generated a syngeneic mouse model of huHER2+ breast cancer, using a truncated form of huHER2, HER2T. Following validation of this model, we next treated tumour-bearing with our immunotherapy strategy: oncolytic vesicular stomatitis virus (VSVΔ51) with clinically approved antibody-drug conjugate targeting huHER2, trastuzumab emtansine (T-DM1). We assessed efficacy through tumour control, survival, and immune analyses.
    The generated truncated HER2T construct was non-immunogenic in wildtype BALB/c mice upon expression in murine mammary carcinoma 4T1.2 cells. Treatment of 4T1.2-HER2T tumours with VSVΔ51+T-DM1 yielded robust curative efficacy compared to controls, and broad immunologic memory. Interrogation of anti-tumour immunity revealed tumour infiltration by CD4+ T cells, and activation of B, NK, and dendritic cell responses, as well as tumour-reactive serum IgG.
    The 4T1.2-HER2T model was used to evaluate the anti-tumour immune responses following our complex pharmacoviral treatment strategy. These data demonstrate utility of the syngeneic HER2T model for assessment of huHER2-targeted therapies in an immune-competent in vivo setting. We further demonstrated that HER2T can be implemented in multiple other syngeneic tumour models, including but not limited to colorectal and ovarian models. These data also suggest that the HER2T platform may be used to assess a range of surface-HER2T targeting approaches, such as CAR-T, T-cell engagers, antibodies, or even retargeted oncolytic viruses.
    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(OVs)是有前途的抗癌治疗方法,可以在癌细胞中特异性复制并杀死癌细胞,并具有深远的免疫刺激作用。我们先前报道了钒基化合物如硫酸氧钒(VS)作为OV免疫疗法的免疫刺激增强剂的潜力。这些化合物,与基于RNA的OV如溶瘤性水泡性口炎病毒(VSVΔ51)结合使用,改善病毒传播和溶瘤,在耐药肿瘤模型中导致长期抗肿瘤免疫和延长生存期。这种效应与在钒存在下病毒诱导的抗病毒I型IFN应答向II型IFN应答转变有关。这里,我们调查了VS+VSVΔ51联合治疗的全身影响,以了解导致抗肿瘤反应改善的免疫学作用机制.VS+VSVΔ51联合治疗显著增加IFN-γ和IL-6的水平,并改善肿瘤抗原特异性T细胞应答。由免疫学分析支持,并作为设计更有效治疗方案的概念证明,我们发现,在同基因CT26WT结肠癌模型中,使用VSVΔ51联合VS的IL-12局部递送可进一步改善治疗结果.
    Oncolytic viruses (OVs) are promising anticancer treatments that specifically replicate in and kill cancer cells and have profound immunostimulatory effects. We previously reported the potential of vanadium-based compounds such as vanadyl sulfate (VS) as immunostimulatory enhancers of OV immunotherapy. These compounds, in conjunction with RNA-based OVs such as oncolytic vesicular stomatitis virus (VSVΔ51), improve viral spread and oncolysis, leading to long-term antitumor immunity and prolonged survival in resistant tumor models. This effect is associated with a virus-induced antiviral type I IFN response shifting towards a type II IFN response in the presence of vanadium. Here, we investigated the systemic impact of VS+VSVΔ51 combination therapy to understand the immunological mechanism of action leading to improved antitumor responses. VS+VSVΔ51 combination therapy significantly increased the levels of IFN-γ and IL-6, and improved tumor antigen-specific T-cell responses. Supported by immunological profiling and as a proof of concept for the design of more effective therapeutic regimens, we found that local delivery of IL-12 using VSVΔ51 in combination with VS further improved therapeutic outcomes in a syngeneic CT26WT colon cancer model.
    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
    阻断磷酸肌醇3-激酶(PI3K)/蛋白激酶B(Akt)信号通路可有效增加溶瘤病毒对癌细胞的细胞毒作用,但是详细的机制仍然未知。基于此,本研究设法研究了PI3K抑制剂ZSTK474和溶瘤性水疱性口炎病毒VSVΔ51联合治疗对骨肉瘤(OS)的体外和体内抗肿瘤作用。具体来说,ZSTK474通过触发内质网(ER)应激介导的凋亡性细胞死亡,加重了VSVΔ51对骨肉瘤发育的抑制作用。机械上,ZSTK474或VSVΔ51单独对骨肉瘤细胞的细胞活力影响有限,而ZSTK474和VSVΔ51联合治疗可显着诱导骨肉瘤细胞凋亡。有趣的是,VSVΔ51增加IRE1α和p-PERK的表达水平,以启动骨肉瘤细胞中的ER应激,通过用ZSTK474共处理细胞而加重。接下来,ZSTK474-VSVΔ51联合治疗对骨肉瘤细胞死亡的促进作用被ER-应激抑制剂4-苯基丁酸(4-PBA)消除,表明ZSTK474以ER-应激依赖性方式增强了VSVΔ51对骨肉瘤细胞的细胞毒性作用。最后,建立小鼠移植瘤模型,结果表明,ZSTK474-VSVΔ51联合治疗可协同抑制体内骨肉瘤细胞的肿瘤发生。一起来看,我们的数据表明,ZSTK474是一种通过加重内质网应激来增强VSVΔ51对骨肉瘤的细胞毒性作用的新药物,本研究可能为骨肉瘤的临床治疗提供替代疗法。
    Blockage of phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signal pathway is effective to increase the cytotoxic effects of oncolytic virus on cancer cells, but the detailed mechanisms are still largely unknown. Based on this, the present study managed to investigate the anti-tumor effects of PI3K inhibitor ZSTK474 and oncolytic vesicular stomatitis virus VSVΔ51 combination treatments on osteosarcoma (OS) in vitro and in vivo. Specifically, ZSTK474 aggravated the inhibiting effects of VSVΔ51 on osteosarcoma development by triggering endoplasmic reticulum (ER)-stress mediated apoptotic cell death. Mechanistically, either ZSTK474 or VSVΔ51 alone had limited effects on cell viability in osteosarcoma cells, while ZSTK474 and VSVΔ51 combination treatments significantly induced osteosarcoma cell apoptosis. Interestingly, VSVΔ51 increased the expression levels of IRE1α and p-PERK to initiate ER stress in osteosarcoma cells, which were aggravated by co-treating cells with ZSTK474. Next, the promoting effects of ZSTK474-VSVΔ51 combined treatment on osteosarcoma cell death were abrogated by the ER-stress inhibitor 4-phenyl butyric acid (4-PBA), indicating that ZSTK474 enhanced the cytotoxic effects of VSVΔ51 on osteosarcoma cells in an ER-stress dependent manner. Finally, the xenograft tumor-bearing mice models were established, and the results showed that ZSTK474-VSVΔ51 combined treatment synergistically hindered tumorigenesis of osteosarcoma cells in vivo. Taken together, our data suggested that ZSTK474 was a novel agent to enhance the cytotoxic effects of VSVΔ51 on osteosarcoma by aggravating ER-stress, and the present study might provide alternative therapy treatments for osteosarcoma in clinic.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号