Oncolytic virotherapy

溶瘤病毒疗法
  • 文章类型: Journal Article
    结直肠癌是世界上第三大常见癌症和第二大致死性癌症。该疾病的主要原因是由于饮食和行为因素。这种复杂疾病的治疗主要以传统治疗为主,包括手术,放射治疗,和化疗。由于其高患病率和高发病率,迫切需要更有效、副作用更少的治疗方法。近年来,免疫疗法已成为潜在的治疗替代方案,也是发展最快的治疗方法之一。免疫疗法通过激活或增强免疫系统以识别和攻击癌细胞来抑制肿瘤生长。这篇综述介绍了免疫检查点抑制剂的最新免疫疗法。细胞疗法,肿瘤浸润淋巴细胞,和溶瘤病毒。其中一些在临床试验中显示出有希望的结果,并用于临床治疗。
    Colorectal cancer is the third most common cancer and the second most lethal cancer in the world. The main cause of the disease is due to dietary and behavioral factors. The treatment of this complex disease is mainly based on traditional treatments, including surgery, radiotherapy, and chemotherapy. Due to its high prevalence and high morbidity, more effective treatments with fewer side effects are urgently needed. In recent years, immunotherapy has become a potential therapeutic alternative and one of the fastest-developing treatments. Immunotherapy inhibits tumor growth by activating or enhancing the immune system to recognize and attack cancer cells. This review presents the latest immunotherapies for immune checkpoint inhibitors, cell therapy, tumor-infiltrating lymphocytes, and oncolytic viruses. Some of these have shown promising results in clinical trials and are used in clinical 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
    背景:神经母细胞瘤是儿童中最常见的颅外实体瘤,占儿科癌症相关死亡的15%。由于肿瘤微环境中免疫细胞的缺乏以及神经母细胞瘤肿瘤细胞释放免疫抑制细胞因子,用免疫疗法靶向神经母细胞瘤已被证明具有挑战性。我们假设将溶瘤性单纯疱疹病毒(oHSV)与自然杀伤(NK)细胞结合可能会克服这些障碍并引起肿瘤细胞死亡。
    方法:我们利用MYCN扩增和非扩增神经母细胞瘤细胞系,表达oHSV的IL-12,M002和人类NK细胞系,NK-92MI。我们评估了NK细胞对有和没有M002感染的神经母细胞瘤的细胞毒性,M002对NK细胞启动的影响,以及M002和引发对NK细胞迁移能力和CD107a表达的影响。为了测试临床适用性,然后我们在体内研究了M002和NK细胞对神经母细胞瘤的影响。
    结果:NK细胞更容易被M002感染的神经母细胞瘤细胞所吸引。M002和NK细胞的联合治疗在体外和体内都增加了神经母细胞瘤细胞死亡。启动NK细胞增强了它们的细胞毒性,迁移能力和CD107a表达。
    结论:据我们所知,这些研究首次证明溶瘤病毒联合自我维持NK细胞在神经母细胞瘤中的作用以及神经母细胞瘤对NK细胞的启动作用.当前的研究提供了对NK细胞与神经母细胞瘤之间关系的更深入理解,这些数据表明oHSV增加了NK细胞对神经母细胞瘤的细胞毒性。
    BACKGROUND: Neuroblastoma is the most common extracranial solid tumor in children and accounts for 15% of pediatric cancer related deaths. Targeting neuroblastoma with immunotherapies has proven challenging due to a paucity of immune cells in the tumor microenvironment and the release of immunosuppressive cytokines by neuroblastoma tumor cells. We hypothesized that combining an oncolytic Herpes Simplex Virus (oHSV) with natural killer (NK) cells might overcome these barriers and incite tumor cell death.
    METHODS: We utilized MYCN amplified and non-amplified neuroblastoma cell lines, the IL-12 expressing oHSV, M002, and the human NK cell line, NK-92 MI. We assessed the cytotoxicity of NK cells against neuroblastoma with and without M002 infection, the effects of M002 on NK cell priming, and the impact of M002 and priming on the migratory capacity and CD107a expression of NK cells. To test clinical applicability, we then investigated the effects of M002 and NK cells on neuroblastoma in vivo.
    RESULTS: NK cells were more attracted to neuroblastoma cells that were infected with M002. There was an increase in neuroblastoma cell death with the combination treatment of M002 and NK cells both in vitro and in vivo. Priming the NK cells enhanced their cytotoxicity, migratory capacity and CD107a expression.
    CONCLUSIONS: To the best of our knowledge, these investigations are the first to demonstrate the effects of an oncolytic virus combined with self-maintaining NK cells in neuroblastoma and the priming effect of neuroblastoma on NK cells. The current studies provide a deeper understanding of the relation between NK cells and neuroblastoma and these data suggest that oHSV increases NK cell cytotoxicity towards neuroblastoma.
    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细胞对实体瘤的成功有限。这里,我们使用溶瘤泡沫病毒(oFV)与抗CD19CART细胞联合在肿瘤上展示模型CAR靶抗原(CD19).我们产生了oFV-Δbel2和oFV-bel2载体以测试病毒/CD19传播的效率和稳定性。虽然这两种病毒在体外具有相同的CAR-T杀伤能力,OFV-Δbel2病毒获得了G到A突变,而oFV-bel2病毒有基因组缺失。在体内皮下肿瘤模型中,CART细胞导致OFV特异性生物发光的显著降低,确认OFV感染的肿瘤细胞的清除。然而,最有效的治疗是在缺乏CAR-T细胞的情况下使用高剂量OFV,表明OFV的CART清除是有害的。此外,在逃避CART细胞治疗的肿瘤中,复发病毒在OFV-CD19转基因中含有缺失,允许病毒逃避CART消除。因此,OFV代表一种缓慢阴燃的溶瘤病毒,其通过肿瘤的慢性扩散产生了抗肿瘤治疗,CART疗法废除了。这些结果表明,该溶瘤平台的进一步发展,有了额外的免疫治疗武器,可能允许慢性溶瘤的有效组合。
    Chimeric antigen receptor (CAR) T cells have had limited success against solid tumors. Here, we used an oncolytic foamy virus (oFV) to display a model CAR target antigen (CD19) on tumors in combination with anti-CD19 CAR T cells. We generated oFV-Δbel2 and oFV-bel2 vectors to test the efficiency and stability of viral/CD19 spread. While both viruses conferred equal CAR T killing in vitro, the oFV-Δbel2 virus acquired G-to-A mutations, whereas oFV-bel2 virus had genome deletions. In subcutaneous tumor models in vivo, CAR T cells led to a significant decrease in oFV-specific bioluminescence, confirming clearance of oFV-infected tumor cells. However, the most effective therapy was with high-dose oFV in the absence of CAR T cells, indicating that CAR T clearance of oFV was detrimental. Moreover, in tumors that escaped CAR T cell treatment, resurgent virus contained deletions within the oFV-CD19 transgene, allowing the virus to escape CAR T elimination. Therefore, oFV represents a slow smoldering type of oncolytic virus, whose chronic spread through tumors generates anti-tumor therapy, which is abolished by CAR T therapy. These results suggest that further development of this oncolytic platform, with additional immunotherapeutic arming, may allow for an effective combination of chronic oncolysis.
    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
    尽管近几十年来黑色素瘤发病率不断上升,有总体死亡率下降的趋势,反映了多种因素,包括改善转移性疾病的治疗方案。虽然局部治疗是早期黑色素瘤的支柱,转移性疾病需要全身治疗,溶瘤病毒疗法成为一种有希望的选择。对于这篇评论,从1964年到2024年从PubMed检索到的文章。我们进行了标题,根据系统审查和荟萃分析指南的首选报告项目进行摘要和全文筛选,以确定描述柯萨奇病毒A21(V937)使用的文章,作为单一疗法或作为恶性黑色素瘤联合疗法的一部分。15篇文章符合纳入标准,提供有关V937在减少肿瘤负担方面的疗效的临床前和临床数据。除了报告可管理的安全概况外,与免疫检查点抑制剂单一疗法相比,检查肿瘤内V937联合治疗的临床试验数据也认可了有利的客观反应率(47%vs.38%和21%vs.10%,分别)。相比之下,尽管在肿瘤组织中达到了可检测的水平(1×109TCID50),但静脉V937单药治疗未能在一组IIIC/IV期黑色素瘤患者(n=3)中产生额外获益.尽管一小部分患者经历了严重的不良反应,并且研究设计限制了收集的数据,V937疗效的证据仍然令人鼓舞.很少有临床试验评估黑色素瘤中的V937,在常规使用转移性病灶的标准治疗之前,还需要额外的数据.
    Despite rising melanoma incidence in recent decades, there is a trend towards overall decreased mortality, reflecting multiple factors including improved treatment options for metastatic disease. While local treatments are the mainstay for early-stage melanoma, metastatic disease necessitates systemic treatment, with oncolytic virotherapy emerging as a promising option. For this review, articles were retrieved from PubMed from 1964 through 2024. We conducted title, abstract and full-text screening in accordance with Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines to identify articles describing the use of coxsackievirus A21 (V937), either as monotherapy or as part of combination therapy for malignant melanoma. Fifteen articles met inclusion criteria, offering preclinical and clinical data on V937\'s efficacy in reducing tumour burden. In addition to reporting manageable safety profiles, clinical trial data examining intratumoral V937 combination therapy with pembrolizumab and ipilimumab also endorsed favourable objective response rates compared to immune checkpoint inhibitor monotherapy (47% vs. 38% and 21% vs. 10%, respectively). In contrast, intravenous V937 monotherapy failed to yield additional benefit in a cohort of patients with Stage IIIC/IV melanoma (n = 3) despite achieving detectable levels in tumour tissue (1 × 109 TCID50). Although small subsets of patients experienced severe adverse effects and study design limitations imposed constraints on collected data, evidence for the efficacy of V937 remains encouraging. With few clinical trials evaluating V937 in melanoma, additional data is required before routine usage in standard treatment for metastatic lesions.
    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
    增殖细胞核抗原(PCNA)是一种有据可查的DNA修复和复制辅助蛋白。它属于围绕DNA的滑动夹蛋白家族,并充当相互作用蛋白的移动对接平台,以安装和执行其代谢任务。PCNA的存在普遍存在于所有细胞中,当位于细胞核中时,它在DNA复制和修复中起作用,细胞周期调控和增殖细胞凋亡。它在某些病毒的感染性中也起着至关重要的作用,如单纯疱疹病毒(HSV)。然而,最近在免疫细胞如嗜中性粒细胞和巨噬细胞的细胞质中发现了它,在那里它已被证明与促炎状态的发展有关。PCNA也在某些癌细胞的表面表达,可以在阻止免疫细胞杀死肿瘤方面发挥作用,以及与癌症毒力有关。鉴于对溶瘤病毒(OVs)作为一种新型癌症治疗的兴趣日益浓厚,这篇综述认为PCNA在健康中的作用,癌变,和免疫细胞,以了解PCNA靶向治疗和溶瘤病毒治疗在未来可能如何相互作用。
    Proliferating cell nuclear antigen (PCNA) is a well-documented accessory protein of DNA repair and replication. It belongs to the sliding clamp family of proteins that encircle DNA and acts as a mobile docking platform for interacting proteins to mount and perform their metabolic tasks. PCNA presence is ubiquitous to all cells, and when located in the nucleus it plays a role in DNA replication and repair, cell cycle control and apoptosis in proliferating cells. It also plays a crucial role in the infectivity of some viruses, such as herpes simplex viruses (HSVs). However, more recently it has been found in the cytoplasm of immune cells such as neutrophils and macrophages where it has been shown to be involved in the development of a pro-inflammatory state. PCNA is also expressed on the surface of certain cancer cells and can play a role in preventing immune cells from killing tumours, as well as being associated with cancer virulence. Given the growing interest in oncolytic viruses (OVs) as a novel cancer therapeutic, this review considers the role of PCNA in healthy, cancerous, and immune cells to gain an understanding of how PCNA targeted therapy and oncolytic virotherapy may interact in the future.
    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
    溶瘤病毒联合免疫疗法在肿瘤治疗中具有重要的潜力。在这项研究中,我们设计了一种进一步减毒的伪狂犬病病毒(PRV)疫苗株,该疫苗株掺入了PD-L1抑制剂,并证明了其在肿瘤治疗中作为溶瘤病毒的前景.我们首先证明了自然减毒PRV疫苗株Bartha可以有效感染来自多个物种的肿瘤细胞,包括人类,老鼠,和狗在体外。然后,我们使用B16-F10黑色素瘤小鼠模型评估了该疫苗株及其不同的单基因缺失突变体的安全性和抗肿瘤功效。TK缺失菌株成为最佳载体,我们使用CRISPR/Cas9技术在其中插入了PD-L1抑制剂(iPD-L1)。与对照相比,重组PRV(rPRV-iPD-L1)在B16-F10黑色素瘤小鼠模型中表现出更显著的抗肿瘤作用.我们的研究表明,PRV不仅可以作为溶瘤病毒开发,而且可以作为表达外源基因以调节肿瘤微环境的强大载体开发。
    Oncolytic viruses combined with immunotherapy offer significant potential in tumor therapy. In this study, we engineered a further attenuated pseudorabies virus (PRV) vaccine strain that incorporates a PD-L1 inhibitor and demonstrated its promise as an oncolytic virus in tumor therapy. We first showed that the naturally attenuated PRV vaccine strain Bartha can efficiently infect tumor cells from multiple species, including humans, mice, and dogs in vitro. We then evaluated the safety and anti-tumor efficacy of this vaccine strain and its different single-gene deletion mutants using the B16-F10 melanoma mouse model. The TK deletion strain emerged as the optimal vector, and we inserted a PD-L1 inhibitor (iPD-L1) into it using CRISPR/Cas9 technology. Compared with the control, the recombinant PRV (rPRV-iPD-L1) exhibited more dramatic anti-tumor effects in the B16-F10 melanoma mouse model. Our study suggests that PRV can be developed not only as an oncolytic virus but also a powerful vector for expressing foreign genes to modulate the tumor microenvironment.
    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(OV)是癌症免疫疗法中一种有前途的策略。它们在局部促进抗肿瘤免疫的能力提高了人们的希望,即对当前免疫疗法无反应的癌症可以更有效地解决。在这种情况下,必须考虑肿瘤相关巨噬细胞(TAM),因为它们在癌症免疫中的关键作用。即使TAM倾向于抑制抗肿瘤反应,它们分泌促炎细胞因子和吞噬癌细胞的能力可用于促进治疗性癌症免疫。OV具有促进有利于抗肿瘤免疫的TAM促炎功能的潜力。但与此同时,TAM促炎功能诱导肿瘤OV清除,从而限制了OV的功效并凸显了OV与TAM的互相感化是一把双刃剑。此外,最近开发了工程化OV来调节特定的TAM功能,例如吞噬活性。在静脉内施用后,循环单核细胞将OV递送到肿瘤中的潜力也正在显现。在这次审查中,我们将介绍OV和TAM之间的相互作用,工程化OV调节特定TAM功能的潜力,以及循环单核细胞在OV递送至肿瘤中的有希望的作用。
    Oncolytic viruses (OV) are a promising strategy in cancer immunotherapy. Their capacity to promote anti-tumoral immunity locally raises hope that cancers unresponsive to current immunotherapy approaches could be tackled more efficiently. In this context, tumor-associated macrophages (TAM) must be considered because of their pivotal role in cancer immunity. Even though TAM tend to inhibit anti-tumoral responses, their ability to secrete pro-inflammatory cytokines and phagocytose cancer cells can be harnessed to promote therapeutic cancer immunity. OVs have the potential to promote TAM pro-inflammatory functions that favor anti-tumoral immunity. But in parallel, TAM pro-inflammatory functions induce OV clearance in the tumor, thereby limiting OV efficacy and highlighting that the interaction between OV and TAM is a double edge sword. Moreover, engineered OVs were recently developed to modulate specific TAM functions such as phagocytic activity. The potential of circulating monocytes to deliver OV into the tumor after intravenous administration is also emerging. In this review, we will present the interaction between OV and TAM, the potential of engineered OV to modulate specific TAM functions, and the promising role of circulating monocytes in OV delivery to the tumor.
    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
    靶向抗肿瘤免疫疗法已取得显著的临床效果。然而,由于靶标缺失或抗原脱落而对这些疗法产生的耐药性限制了其疗效,并将肿瘤排除在候选之外.为了解决这个限制,在这里,我们设计了一种溶瘤弹状病毒,水泡性口炎病毒(VSVΔ51),表达截短的靶向抗原,这允许用曲妥珠单抗靶向HER2。截短的HER2(HER2T)缺乏信号传导能力并且在感染的细胞表面上有效表达。VSVΔ51介导的HER2T表达模拟肿瘤中HER2阳性状态,使抗体-药物缀合物曲妥珠单抗在体外有效治疗,离体,和体内。此外,我们将VSVΔ51-HER2T与表达HER2靶向T细胞衔接剂的溶瘤痘苗病毒组合。这种双病毒治疗策略在雌性小鼠中使用CD3+浸润物进行抗肿瘤免疫表现出有效的体内疗效。我们的发现展示了使用溶瘤病毒定制肿瘤微环境的能力,从而增强与“现成”靶向疗法的兼容性。
    Targeted antineoplastic immunotherapies have achieved remarkable clinical outcomes. However, resistance to these therapies due to target absence or antigen shedding limits their efficacy and excludes tumours from candidacy. To address this limitation, here we engineer an oncolytic rhabdovirus, vesicular stomatitis virus (VSVΔ51), to express a truncated targeted antigen, which allows for HER2-targeting with trastuzumab. The truncated HER2 (HER2T) lacks signaling capabilities and is efficiently expressed on infected cell surfaces. VSVΔ51-mediated HER2T expression simulates HER2-positive status in tumours, enabling effective treatment with the antibody-drug conjugate trastuzumab emtansine in vitro, ex vivo, and in vivo. Additionally, we combine VSVΔ51-HER2T with an oncolytic vaccinia virus expressing a HER2-targeted T-cell engager. This dual-virus therapeutic strategy demonstrates potent curative efficacy in vivo in female mice using CD3+ infiltrate for anti-tumour immunity. Our findings showcase the ability to tailor the tumour microenvironment using oncolytic viruses, thereby enhancing compatibility with \"off-the-shelf\" targeted therapies.
    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
    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
    弹状病毒载体可以诱导癌细胞的裂解。虽然几乎只在37°C下进行研究,病毒在体内会受到一系列温度的影响,包括温度≤31℃。尽管有潜在的影响,温度<37°C对弹状病毒载体性能的影响尚不清楚。我们研究了低解剖温度对两种弹状病毒的影响,水泡性口炎病毒(VSV)和马拉巴病毒(MG1)。使用新陈代谢的刃天青测定,在28、31、34和37°C的一组细胞系中表征了VSV和MG1介导的溶瘤作用。两种病毒的溶瘤能力在31和28°C都受到阻碍。尝试了两种病毒的冷适应作为缓解策略。在降低的温度下连续传代病毒以试图诱导突变。不幸的是,冷适应策略未能在<37°C的温度下增强病毒的溶瘤活性。有趣的是,我们发现,尽管溶瘤活性被消除,但在低温下病毒复制不受影响.相比之下,癌细胞的增殖在低温下减少。如果将低温下的细胞用病毒处理更长的时间,则可以实现等效的溶瘤作用。这表明弹状病毒介导的溶瘤作用可能在治疗窗口有限的体内低温下受损。
    Rhabdoviral vectors can induce lysis of cancer cells. While studied almost exclusively at 37 °C, viruses are subject to a range of temperatures in vivo, including temperatures ≤31 °C. Despite potential implications, the effect of temperatures <37 °C on the performance of rhabdoviral vectors is unknown. We investigated the effect of low anatomical temperatures on two rhabdoviruses, vesicular stomatitis virus (VSV) and Maraba virus (MG1). Using a metabolic resazurin assay, VSV- and MG1-mediated oncolysis was characterized in a panel of cell lines at 28, 31, 34 and 37 °C. The oncolytic ability of both viruses was hindered at 31 and 28 °C. Cold adaptation of both viruses was attempted as a mitigation strategy. Viruses were serially passaged at decreasing temperatures in an attempt to induce mutations. Unfortunately, the cold-adaptation strategies failed to potentiate the oncolytic activity of the viruses at temperatures <37 °C. Interestingly, we discovered that viral replication was unaffected at low temperatures despite the abrogation of oncolytic activity. In contrast, the proliferation of cancer cells was reduced at low temperatures. Equivalent oncolytic effects could be achieved if cells at low temperatures were treated with viruses for longer times. This suggests that rhabdovirus-mediated oncolysis could be compromised at low temperatures in vivo where therapeutic windows are limited.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号