oHSV

oHSV
  • 文章类型: Journal Article
    溶瘤病毒(OVs)是一种新兴的肿瘤免疫方法,其允许使用天然或遗传修饰的病毒特异性感染和裂解肿瘤细胞而不损害正常细胞。溶瘤性单纯疱疹病毒(oHSV)是肿瘤学领域研究和应用较为广泛的OVs,可以直接杀死肿瘤细胞,促进抗肿瘤免疫反应。oHSV是少数具有良好抗病毒药物的病毒之一,所以oHSV在临床上也更安全。近年来,除了肿瘤中oHSV的单一疗法外,越来越多的研究致力于探索oHSV联合其他治疗方法的抗肿瘤作用。在这篇文章中,我们描述了oHSV联合治疗神经系统肿瘤的进展,消化系统,生殖系统和其他系统。
    Oncolytic Virus (OVs) is an emerging approach to tumour immunity that allows the use of natural or genetically modified viruses to specifically infect and lyse tumour cells without damaging normal cells. Oncolytic herpes simplex virus (oHSV) is one of the more widely researched and applied OVs in the field of oncology, which can directly kill tumour cells to promote anti-tumour immune responses. oHSV is one of the few viruses with good antiviral drugs, so oHSV is also more clinically safe. In recent years, in addition to monotherapy of oHSV in tumours, more and more studies have been devoted to exploring the anti-tumour effects of oHSV in combination with other therapeutic approaches. In this article we describe the progress of oHSV combination therapy against tumours in the nervous system, digestive system, reproductive system and other systems.
    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
    能够在感染细胞中形成合胞体的病毒修饰可以增强溶瘤性单纯疱疹病毒(oHSV)的裂解,该病毒选择性地杀死癌细胞。在受体重新靶向的oHSV(RR-oHSV)的情况下,它只能进入并扩散到癌细胞,通过将病毒修饰为合胞体类型(RRsyn-oHSV),可以将抗肿瘤作用增强至>100,000倍。然而,当包含非癌细胞的合胞体通过条件复制合胞体oHSV(CRsyn-oHSV)诱导时,合胞体死亡发生在早期。这导致CRsyn-oHSV的抗肿瘤作用有限。这里,我们研究了坏死是否与癌细胞和非癌细胞融合形成的合胞体的死亡有关.混合谱系激酶结构域样(MLKL),执行坏死的分子,在所有被检查的鼠癌细胞系中表达,而受体相互作用蛋白激酶3(RIPK3),磷酸化MLKL,在大多数细胞系中都不存在。相比之下,RIPK3在非癌鼠成纤维细胞系中表达。当CRsyn-oHSV感染的RIPK3缺陷癌细胞系与成纤维细胞细胞系共培养时,但不是癌细胞本身,MLKL磷酸化并诱导合胞体死亡。这些结果表明,当CRsyn-oHSV还包含非癌细胞时,在由CRsyn-oHSV形成的多核巨细胞中诱导了早期坏死。
    Viral modifications enabling syncytium formation in infected cells can augment lysis by oncolytic herpes simplex viruses (oHSVs) which selectively kill cancer cells. In the case of receptor-retargeted oHSVs (RR-oHSVs) that exclusively enter and spread to cancer cells, anti-tumor effects can be enhanced in a magnitude of >100,000-fold by modifying the virus to a syncytial type (RRsyn-oHSV). However, when syncytia containing non-cancerous cells are induced by conditionally replicating syncytial oHSV (CRsyn-oHSV), syncytial death occurs at an early stage. This results in limited anti-tumor effects of the CRsyn-oHSV. Here, we investigated whether necroptosis is involved in death of the syncytia formed by the fusion of cancer cells and non-cancerous cells. Mixed-lineage kinase domain-like (MLKL), a molecule executing necroptosis, was expressed in all murine cancer cell lines examined, while receptor-interacting protein kinase 3 (RIPK3), which phosphorylates MLKL, was absent from most cell lines. In contrast, RIPK3 was expressed in non-cancerous murine fibroblast cell lines. When a CRsyn-oHSV-infected RIPK3-deficient cancer cell line was co-cultured with the fibroblast cell line, but not with the cancer cells themselves, MLKL was phosphorylated and syncytial death was induced. These results indicate that early necroptosis is induced in multinucleated giant cells formed by CRsyn-oHSV when they also contain non-cancerous 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
    胶质母细胞瘤是最常见和侵袭性的恶性脑肿瘤,治疗选择有限。因此,迫切需要创新的方法。溶瘤病毒疗法由于其肿瘤特异性靶向和免疫刺激特性而成为癌症治疗的有希望的方式。在这项研究中,我们通过删除15kb的内部重复区和γ34.5基因的两个拷贝,开发了新一代的溶瘤性单纯疱疹病毒C5252。此外,C5252配备了抗程序性细胞死亡蛋白1抗体和白细胞介素12,以增强其对胶质母细胞瘤免疫病毒疗法的治疗功效。体外和体内实验表明,C5252具有显着的安全性和针对胶质母细胞瘤的有效抗肿瘤活性。机制研究表明,C5252通过下调睫状神经营养因子受体α,通过caspase-3/7激活特异性诱导细胞凋亡。此外,证实了C5252在皮下胶质母细胞瘤模型和原位胶质母细胞瘤模型中的抗肿瘤疗效增强.此外,同基因小鼠模型表明,与无武装的骨干病毒相比,C5252的鼠替代物具有优越的抗肿瘤活性,增强的免疫激活。一起来看,我们的发现支持C5252作为胶质母细胞瘤治疗的一个有希望的治疗选择,将其定位为临床翻译的非常有希望的候选人。
    Glioblastoma is the most common and aggressive malignant brain tumor and has limited treatment options. Hence, innovative approaches are urgently needed. Oncolytic virus therapy is emerging as a promising modality for cancer treatment due to its tumor-specific targeting and immune-stimulatory properties. In this study, we developed a new generation of oncolytic herpes simplex virus C5252 by deletion of a 15-kb internal repeat region and both copies of γ34.5 genes. Additionally, C5252 was armed with anti-programmed cell death protein 1 antibody and interleukin-12 to enhance its therapeutic efficacy for glioblastoma immune-virotherapy. In vitro and in vivo experiments demonstrate that C5252 has a remarkable safety profile and potent anti-tumor activity against glioblastoma. Mechanistic studies demonstrated that C5252 specifically induces cell apoptosis by caspase-3/7 activation via downregulating ciliary neurotrophic factor receptor α. Furthermore, the enhanced anti-tumor therapeutic efficacy of C5252 in a subcutaneous glioblastoma model and an orthotopic glioblastoma model was confirmed. Moreover, syngeneic mouse models showed that the murine surrogate of C5252 has superior anti-tumor activity compared to the unarmed backbone virus, with enhanced immune activation. Taken together, our findings support C5252 as a promising therapeutic option for glioblastoma treatment, positioning it as a highly promising candidate for clinical translation.
    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(GBM)是最致命的癌症之一,使用标准治疗(手术,辐射,和化疗),几十年来没有显著延长。GBM表现出显著的细胞异质性,先端有胶质母细胞瘤干细胞样细胞(GSC)。GSC是具有自我更新能力的GBM细胞亚群,区分,启动肿瘤形成,并操纵肿瘤微环境(TME)。GSC不再被认为是具有特异性标志物的静态细胞群,但在表型上和驱动肿瘤异质性和治疗抗性方面是相当灵活的。鉴于这些特点,它们是GBM治疗成功的关键目标。溶瘤病毒,特别是溶瘤单纯疱疹病毒(oHSV),具有许多治疗属性,并且是靶向GSC的有前途的药物。oHSV经过基因工程改造,可以选择性地在癌细胞中复制并杀死癌细胞,包括GSC,但不是正常细胞。此外,oHSV可以诱导抗肿瘤免疫反应,并与其他疗法协同作用,比如化疗,DNA修复抑制剂,免疫检查点抑制剂,以增强治疗效果并减少部分负责化学和放射性抗性的GSC群体。在这里,我们概述了GSC,不同OHSV的活动,临床试验结果,以及提高疗效的组合策略,包括OHSV的治疗武装。在整个过程中,治疗重点将放在GSCs和专门针对这些细胞的研究上.最近的临床试验和oHSVG47Δ在日本对复发性神经胶质瘤患者的批准证明了oHSV治疗的有效性和前景。
    Glioblastoma (GBM) is one of the most lethal cancers, having a poor prognosis and a median survival of only about 15 months with standard treatment (surgery, radiation, and chemotherapy), which has not been significantly extended in decades. GBM demonstrates remarkable cellular heterogeneity, with glioblastoma stem-like cells (GSCs) at the apex. GSCs are a subpopulation of GBM cells that possess the ability to self-renew, differentiate, initiate tumor formation, and manipulate the tumor microenvironment (TME). GSCs are no longer considered a static population of cells with specific markers but are quite flexible phenotypically and in driving tumor heterogeneity and therapeutic resistance. In light of these features, they are a critical target for successful GBM therapy. Oncolytic viruses, in particular oncolytic herpes simplex viruses (oHSVs), have many attributes for therapy and are promising agents to target GSCs. oHSVs are genetically-engineered to selectively replicate in and kill cancer cells, including GSCs, but not normal cells. Moreover, oHSV can induce anti-tumor immune responses and synergize with other therapies, such as chemotherapy, DNA repair inhibitors, and immune checkpoint inhibitors, to potentiate treatment effects and reduce GSC populations that are partly responsible for chemo- and radio-resistance. Herein, we present an overview of GSCs, activity of different oHSVs, clinical trial results, and combination strategies to enhance efficacy, including therapeutic arming of oHSV. Throughout, the therapeutic focus will be on GSCs and studies specifically targeting these cells. Recent clinical trials and approval of oHSV G47Δ in Japan for patients with recurrent glioma demonstrate the efficacy and promise of oHSV 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
    高迁移率族蛋白1(HMGB1)是一种损伤相关分子模式(DAMP)分子,在炎症和肿瘤发生中起重要作用。晚期糖基化终产物受体(RAGE)是细胞外HMGB1结合介导其活性的主要受体之一。RAGE在内皮细胞(EC)上高度表达,并在炎症过程中调节内皮通透性。这里,我们将RAGE的内源性分泌形式(esRAGE)作为RAGE配体的诱饵受体引入溶瘤性单纯疱疹病毒1(oHSV)(OVesRAGE),which,释放后,可以阻止RAGE信令。OVesRAGE在体外和体内显着降低了胶质母细胞瘤(GBM)细胞中MEK1/2和Erk的磷酸化,并增加了切割的PARP。使用与EC共培养的oHSV感染的GBM细胞来测试OVesRAGE对EC活化的影响,容器泄漏,病毒复制,和肿瘤细胞杀伤。OVesRAGE可以有效分泌esRAGE并挽救病毒诱导的EC迁移和激活。当与EC共培养时,降低的EC活化促进了病毒在肿瘤细胞中的复制。最后,OVesRAGE显著增强了携带GBM的小鼠的治疗功效。总的来说,我们的数据表明,HMGB1-RAGE信号传导可能是一个有前景的靶标,其抑制是提高oHSV治疗疗效的可行方法.
    High-mobility group box 1 (HMGB1) is a damage-associated molecular pattern (DAMP) molecule that plays an important role in inflammation and tumorigenesis. Receptor for advanced glycation end products (RAGE) is one of the major receptors to which extracellular HMGB1 binds to mediate its activity. RAGE is highly expressed on the endothelial cells (ECs) and regulates endothelial permeability during inflammation. Here, we introduced the endogenous secretory form of RAGE (esRAGE) as a decoy receptor for RAGE ligands into an oncolytic herpes simplex virus 1 (oHSV) (OVesRAGE), which, upon release, can function to block RAGE signaling. OVesRAGE significantly decreased phosphorylation of MEK1/2 and Erk and increased cleaved PARP in glioblastoma (GBM) cells in vitro and in vivo. oHSV-infected GBM cells co-cultured with ECs were used to test OVesRAGE effect on EC activation, vessel leakiness, virus replication, and tumor cell killing. OVesRAGE could effectively secrete esRAGE and rescue virus-induced EC migration and activation. Reduced EC activation facilitated virus replication in tumor cells when co-cultured with ECs. Finally, OVesRAGE significantly enhanced therapeutic efficacy in GBM-bearing mice. Collectively, our data demonstrate that HMGB1-RAGE signaling could be a promising target and that its inhibition is a feasible approach to improve the efficacy of oHSV 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
    背景:已广泛探索溶瘤病毒(OV)的联合疗法,因为它可以抵抗肿瘤。HDAC抑制剂(HDACi)panobinostat是一种有效的全脱乙酰酶抑制剂,可阻断多种癌症相关途径并逆转癌症进展中的表观遗传事件。
    方法:在本研究中,研究了与oHSV和panobinostat联合使用时的体外溶瘤活性和体内抗肿瘤疗效。
    结果:(1)在人神经胶质瘤A172和鳞状细胞癌SCC9细胞中,使用帕比司他治疗可增强oHSV的增殖和细胞毒性。(2)oHSV和panobinostat联合治疗可增强人胶质瘤A172和鳞状细胞癌SCC9细胞中IFN-β和IFN应答抗病毒基因的转录下调介导的病毒复制。(3)Panobinostat治疗可诱导胶质瘤和鳞状细胞癌细胞中PD-L1的表达上调。(4)当用oHSV组合治疗时,鼠神经胶质瘤CT-2A和鳞状细胞癌SCC7模型在体内显示出显著增强的治疗效果,包括PD-1/PD-L1阻断和HDAC抑制。
    结论:因此,这些数据为OVs联合治疗的临床发展提供了一些新的线索,表观遗传修饰剂,以及神经胶质瘤和鳞状细胞癌的检查点封锁。
    Combination therapy has been widely explored for oncolytic virus (OV), as it can be met with tumor resistance. The HDAC inhibitor (HDACi) panobinostat is a potent pan-deacetylase inhibitor which blocks multiple cancer-related pathways and reverses epigenetic events in cancer progression.
    In this study, oncolytic activity in vitro and antitumor therapeutic efficacy in vivo when combined with oHSV and panobinostat were investigated.
    (1) Treatment with panobinostat enhanced oHSV propagation and cytotoxicity in human glioma A172 and squamous cell carcinoma SCC9 cells. (2) Combined treatment with oHSV and panobinostat enhanced virus replication mediated by the transcriptional downregulation of IFN-β- and IFN-responsive antiviral genes in human glioma A172 and squamous cell carcinoma SCC9 cells. (3) Panobinostat treatment induced upregulation of PD-L1 expression in both glioma and squamous cell carcinoma cells. (4) A significantly enhanced therapeutic efficacy was shown in vivo for the murine glioma CT-2A and squamous cell carcinoma SCC7 models when treated with a combination of oHSV, including PD-1/PD-L1 blockade and HDAC inhibition.
    Consequently, these data provide some new clues for the clinical development of combination therapy with OVs, epigenetic modifiers, and checkpoint blockades for glioma and squamous cell carcinoma.
    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
    未经证实:CAR-T细胞疗法是治疗血液系统恶性肿瘤的一种有希望的方法。然而,在实体瘤中的应用仍然面临许多严峻的挑战,包括抗原表达的异质性和免疫抑制肿瘤微环境(TME)。作为一种新的癌症治疗模式,溶瘤病毒疗法可以被设计为规避实体肿瘤中CART细胞疗法的这些障碍。
    未经批准:在这项研究中,OHSVT7011被设计为驱动双抗原的异位表达,CD19和BCMA的细胞外结构域,在实体瘤细胞表面上被批准的CART细胞靶向。此外,多种免疫调节剂,还包括CCL5、IL-12和抗PD-1抗体以调节TME。在体外和体内评估了T7011与CD19或BCMACART细胞组合的抗肿瘤活性。
    UNASSIGNED:T7011感染后可检测到肿瘤细胞表面CD19或BMCA的表达。TME中的CCL5水平也升高。功效研究表明,与T7011和CAR-TCD19或CAR-TBCMA细胞的组合在几种实体瘤模型中显示出显著的协同抗肿瘤应答。
    UNASSIGNED:这些研究表明,新一代oHSVT7011可以是一种有前途的与CD19或BCMA特异性CART细胞的组合疗法,用于治疗广泛的实体瘤。
    UNASSIGNED: The CAR T-cell therapy is a promising approach to treating hematologic malignancies. However, the application in solid tumors still has many tough challenges, including heterogenicity in antigen expressions and immunosuppressive tumor microenvironment (TME). As a new cancer treatment modality, oncolytic virotherapy can be engineered to circumvent these obstacles for CAR T cell therapy in solid tumors.
    UNASSIGNED: In this study, an oHSV T7011 is engineered to drive ectopic expression of dual-antigens, extracellular domains of CD19 and BCMA, on the solid tumor cell surface to be targeted by approved CAR T cells. In addition, multiple immunomodulators, CCL5, IL-12, and anti-PD-1 antibody are also included to modulate the TME. The antitumor activities of T7011 in combination with CD19 or BCMA CAR T-cell were evaluated in vitro and in vivo.
    UNASSIGNED: The expression of CD19 or BMCA on the tumor cell surface could be detected after T7011 infection. The level of CCL5 in TME was also increased. Efficacy studies demonstrated that combination with T7011 and CAR-TCD19 or CAR-TBCMA cells showed significant synergistic anti-tumor responses in several solid tumor models.
    UNASSIGNED: These studies indicated that the new generation of oHSV T7011 can be a promising combinational therapy with CD19 or BCMA-specific CAR T cells for the treatment of a broad range of solid 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
    高级别神经胶质瘤(HGG)是致命的中枢神经系统肿瘤,会在大脑中迅速扩散,使治疗具有挑战性。整合素是介导细胞-细胞外基质(ECM)相互作用的跨膜受体,细胞粘附,迁移,增长,和生存。它们在HGG恶性肿瘤中的上调和负相关使靶向整合素成为可行的治疗选择。整合素也在单纯疱疹病毒1(HSV-1)进入中起作用。溶瘤HSV-1(oHSV)是临床上最先进的溶瘤病毒疗法,显示出优于实体癌标准癌症治疗的安全性和有效性,包括HGG。随着FDA批准用于黑色素瘤的oHSV以及最近在日本有条件批准用于恶性神经胶质瘤的oHSV,将oHSV用于HGG已经引起了极大的兴趣。在这次审查中,我们对整合素在oHSV中的作用进行了系统的概述,特别关注其对HGG的治疗潜力。我们讨论了在oHSV治疗过程中靶向整合素的利弊:虽然整合素通过充当oHSV进入的门户而发挥着促进治疗的作用,它们还介导阻碍oHSV治疗功效的先天抗病毒免疫反应。我们进一步讨论了在oHSV治疗背景下调节整合素双重功能的替代策略。
    High-grade gliomas (HGGs) are lethal central nervous system tumors that spread quickly through the brain, making treatment challenging. Integrins are transmembrane receptors that mediate cell-extracellular matrix (ECM) interactions, cellular adhesion, migration, growth, and survival. Their upregulation and inverse correlation in HGG malignancy make targeting integrins a viable therapeutic option. Integrins also play a role in herpes simplex virus 1 (HSV-1) entry. Oncolytic HSV-1 (oHSV) is the most clinically advanced oncolytic virotherapy, showing a superior safety and efficacy profile over standard cancer treatment of solid cancers, including HGG. With the FDA-approval of oHSV for melanoma and the recent conditional approval of oHSV for malignant glioma in Japan, usage of oHSV for HGG has become of great interest. In this review, we provide a systematic overview of the role of integrins in relation to oHSV, with a special focus on its therapeutic potential against HGG. We discuss the pros and cons of targeting integrins during oHSV therapy: while integrins play a pro-therapeutic role by acting as a gateway for oHSV entry, they also mediate the innate antiviral immune responses that hinder oHSV therapeutic efficacy. We further discuss alternative strategies to regulate the dual functionality of integrins in the context of oHSV 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
    单纯疱疹病毒1型(HSV-1)是唯一被FDA和EMA批准的溶瘤病毒,因此,许多潜在的溶瘤HSV(oHSV)正在临床开发中。利用的oHSV亲本菌株是,然而,主要基于实验室参考菌株,与HSV-1的循环菌株相比,其可能具有受损的细胞溶解能力。这里,我们评估了来自芬兰的36种循环HSV-1菌株的表型,以揭示它们作为oHSV骨架的潜力.首先,我们确定了它们的细胞间扩散能力和细胞间扩散能力,找到具有对每种应用有利的复制谱的菌株。第二,来展现差异,我们研究了两种相关病毒糖蛋白(gB/UL27,gI/US7)的遗传多样性。第三,我们检查了代表神经胶质瘤的细胞中菌株的溶瘤潜力,淋巴瘤和结直肠腺癌。我们的结果表明,循环分离株的表型,包括溶瘤潜能,与宿主细胞类型高度相关。然而,我们在许多或所有研究的癌细胞类型中鉴定出与参考病毒相比具有更高的溶瘤潜能的分离株.我们的研究强调在早期载体设计中需要仔细选择骨干病毒,并强调了临床分离株在oHSV发展中作为骨干的潜力。
    Herpes simplex virus type 1 (HSV-1) is the only FDA- and EMA- approved oncolytic virus, and accordingly, many potential oncolytic HSVs (oHSV) are in clinical development. The utilized oHSV parental strains are, however, mostly based on laboratory reference strains, which may possess a compromised cytolytic capacity in contrast to circulating strains of HSV-1. Here, we assess the phenotype of thirty-six circulating HSV-1 strains from Finland to uncover their potential as oHSV backbones. First, we determined their capacity for cell-to-cell versus extracellular spread, to find strains with replication profiles favorable for each application. Second, to unfold the differences, we studied the genetic diversity of two relevant viral glycoproteins (gB/UL27, gI/US7). Third, we examined the oncolytic potential of the strains in cells representing glioma, lymphoma, and colorectal adenocarcinoma. Our results suggest that the phenotype of a circulating isolate, including the oncolytic potential, is highly related to the host cell type. Nevertheless, we identified isolates with increased oncolytic potential in comparison with the reference viruses across many or all of the studied cancer cell types. Our research emphasizes the need for careful selection of the backbone virus in early vector design, and it highlights the potential of clinical isolates as backbones in oHSV development.
    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
    溶瘤病毒(OV)作为一种有前途的治疗剂,可以选择性地感染和杀死具有天然遗传或工程特性的肿瘤细胞。考虑到OVs单药治疗的局限性,联合治疗已被广泛探索。MEK抑制剂(MEKi)Trametinib是FDA批准的激酶抑制剂,用于治疗具有BRAFV600E或V600K突变的肿瘤。在这项研究中,研究了联合使用oHSV和MEKiTrametinib时的体外溶瘤活性和体内抗肿瘤治疗效果.我们发现:(1)用MEKi曲美替尼治疗可增强BRAFV600E突变肿瘤细胞中的oHSV溶瘤活性。(2)在BRAFV600E突变的肿瘤细胞以及BRAFwt/KRAS突变的肿瘤细胞中,用oHSV和MEKi曲美替尼联合治疗增强了通过下调STAT1和PKR表达或磷酸化介导的病毒复制。(3)BRAFwt/KRAS突变的肿瘤模型在体内显示出明显的协同治疗效果,当oHSV的组合包括PD-1阻断和MEK抑制。总的来说,这些数据为临床开发溶瘤病毒联合治疗提供了一些新的见解,MEK抑制,和BRAF或KRAS突变肿瘤的检查点阻断。
    Oncolytic virus (OV) as a promising therapeutic agent can selectively infect and kill tumor cells with naturally inherited or engineered properties. Considering the limitations of OVs monotherapy, combination therapy has been widely explored. MEK inhibitor (MEKi) Trametinib is an FDA-approved kinase inhibitor indicated for the treatment of tumors with BRAF V600E or V600K mutations. In this study, the oncolytic activity in vitro and anti-tumor therapeutic efficacy in vivo when combined with oHSV and MEKi Trametinib were investigated. We found: (1) Treatment with MEKi Trametinib augmented oHSV oncolytic activity in BRAF V600E-mutated tumor cells. (2) Combination treatment with oHSV and MEKi Trametinib enhanced virus replication mediated by down-regulation of STAT1 and PKR expression or phosphorylation in BRAF V600E-mutated tumor cells as well as BRAF wt/KRAS-mutated tumor cells. (3) A remarkably synergistic therapeutic efficacy was shown in vivo for BRAF wt/KRAS-mutated tumor models, when a combination of oHSV including PD-1 blockade and MEK inhibition. Collectively, these data provide some new insights for clinical development of combination therapy with oncolytic virus, MEK inhibition, and checkpoint blockade for BRAF or KRAS-mutated tumors.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号