oncolytic virotherapy

溶瘤病毒疗法
  • 文章类型: Journal Article
    溶瘤病毒,特别是麻疹病毒,代表了一种有趣的治疗方法,用于预后不良且对常规疗法经常耐药的肿瘤。犬组织细胞肉瘤(HS)是需要新治疗方法的此类肿瘤的例证。先前的研究表明,在异种移植的犬组织细胞肉瘤细胞(DH82细胞)上急性肿瘤内应用犬瘟热病毒(CDV)的成功有限。而持续CDV感染的DH82细胞移植表现出完全自发的消退。因此,本研究的重点是在小鼠模型中将持续的CDV疫苗株Onderstepoort感染的DH82(DH82Ondp.i.)细胞瘤内应用到未感染的皮下DH82细胞移植中。接受10份申请的DH82细胞移植,相隔两天,显示了短暂的生长迟缓以及肿瘤内坏死的较大区域,较低的有丝分裂率,与对照组相比,肿瘤内血管形成减少。在施用DH82Ondp.i.细胞后直到最后一次注射后66天在所有肿瘤中检测到病毒mRNA。此外,感染性病毒一直存在到最后一次注射后62天。虽然没有实现完全回归,本申请方案提供了有希望的结果作为进一步治疗的基础,特别是转基因病毒,以增强观察到的效果。
    Oncolytic viruses and morbilliviruses in particular, represent an interesting therapeutic approach for tumors with a poor prognosis and frequent resistance to conventional therapies. Canine histiocytic sarcomas (HS) exemplify such a neoplasm in need for new curative approaches. Previous investigations demonstrated a limited success of an acute intratumoral application of canine distemper virus (CDV) on xenotransplanted canine histiocytic sarcoma cells (DH82 cells), while persistently CDV-infected DH82 cell transplants exhibited a complete spontaneous regression. Therefore, the present study focuses on an intratumoral application of persistently CDV vaccine strain Onderstepoort-infected DH82 (DH82 Ond p.i.) cells into non-infected subcutaneous DH82 cell transplants in a murine model. DH82 cell transplants that received 10 applications, two days apart, showed a transient growth retardation as well as larger areas of intratumoral necrosis, lower mitotic rates, and a decreased intratumoral vascularization compared to controls. Viral mRNA was detected in all neoplasms following application of DH82 Ond p.i. cells until 66 days after the last injection. Furthermore, infectious virus was present until 62 days after the last injection. Although complete regression was not achieved, the present application regimen provides promising results as a basis for further treatments, particularly with genetically modified viruses, to enhance the observed effects.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    炎性肌纤维母细胞瘤(IMT)是一种罕见的病理实体,最早于1939年描述。这种病变最常见于肺部,但是涉及其他系统的案件,例如称为颅内IMT(IIMT)的中枢神经系统,也有报道。由于缺乏特征性的影像学变化,目前的诊断依赖于病理结果。手术切除是一种有效的治疗方法,虽然这种疾病是侵入性的,可能会复发。以前的文献报道了IMT组织中高水平的程序性死亡1(PD-1)表达,表明免疫疗法可能对这种情况有效。在这个案例报告中,我们介绍了一名中年男性,他在IIMT切除手术后接受了PD-1抑制剂和溶瘤腺病毒(Ad-TD-nsIL12)治疗.这种成功的方法为IIMT的治疗提供了新的方向。
    Inflammatory myofibroblastic tumor (IMT) is a rare pathological entity first described in 1939. This lesion is most commonly found in the lungs, but cases involving other systems, such as the central nervous system known as intracranial IMT (IIMT), have also been reported. Diagnosis currently relies on pathological results due to the lack of characteristic imaging changes. Surgical resection is an effective treatment, though the disease is invasive and may recur. Previous literature has reported a high level of programmed death 1 (PD-1) expression in IMT tissues, suggesting that immunotherapy may be effective for this condition. In this case report, we present a middle-aged male who received PD-1 inhibitor and oncolytic adenovirus (Ad-TD-nsIL12) treatment after IIMT resection surgery. This successful approach provides a new direction for the treatment of IIMT.
    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
    骨肉瘤(OS),儿童和青少年骨最常见的原发性恶性肿瘤,免疫检查点抑制剂由于其抗肿瘤免疫应答不良而难以治疗。化疗和病毒疗法诱导免疫原性细胞死亡(ICD)和抗肿瘤免疫反应,在未经治疗的肿瘤中导致外镜效应。我们先前证明了端粒酶特异性复制型溶瘤腺病毒OBP-301和p53武装的OBP-702在人类OS细胞中的抗肿瘤活性。这里,我们展示了化疗药物(阿霉素,顺铂)和端粒酶特异性溶瘤腺病毒(OBP-301,p53武装的OBP-702)在人OS细胞中诱导ICD(U2OS,MNNG/HOS,SaOS-2)和鼠OS细胞(NHOS)。与化疗和OBP-301相比,OBP-702通过分泌三磷酸腺苷(ATP)和高迁移率族蛋白盒B1(HMGB1)在人OS细胞中诱导更深入的ICD。鼠NHOS细胞对OBP-702也比OBP-301更敏感。皮下NHOS肿瘤模型表明,与OBP-301相比,瘤内注射OBP-702显着增加了细胞毒性CD8T细胞的肿瘤浸润,并诱导了对未治疗肿瘤的外视作用。我们的结果表明,OBP-702是一种有前途的抗肿瘤试剂,可诱导ICD分泌ATP和HMGB1,并具有抗OS的作用。
    Osteosarcoma (OS), the most frequent primary malignant tumor of bone in children and adolescents, is refractory to immune checkpoint inhibitors due to its poor antitumor immune response. Chemotherapy and virotherapy induce immunogenic cell death (ICD) and antitumor immune responses, leading to the abscopal effect in untreated tumors. We previously demonstrated the antitumor activity of the telomerase-specific replication-competent oncolytic adenoviruses OBP-301 and p53-armed OBP-702 in human OS cells. Here, we show the therapeutic potential of chemotherapeutic drugs (doxorubicin, cisplatin) and telomerase-specific oncolytic adenoviruses (OBP-301, p53-armed OBP-702) to induce ICD in human OS cells (U2OS, MNNG/HOS, SaOS-2) and murine OS cells (NHOS). OBP-702 induced more profound ICD via the secretion of adenosine triphosphate (ATP) and high-mobility group box protein B1 (HMGB1) compared with chemotherapy and OBP-301 in human OS cells. Murine NHOS cells were also more sensitive to OBP-702 than OBP-301. Subcutaneous NHOS tumor models demonstrated that intratumoral injection of OBP-702 significantly increased the tumor infiltration of cytotoxic CD8+ T cells and induced the abscopal effect against non-treated tumors compared with OBP-301. Our results suggest that OBP-702 is a promising antitumor reagent to induce ICD with secretion of ATP and HMGB1 and the abscopal effect against OS.
    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
    癌症免疫疗法的使用并不新颖,但已在临床上使用了数十年。直到最近,我们才发现在检查点抑制剂突破后刺激抗肿瘤反应的真正潜力。癌症免疫疗法已成为许多不同阶段恶性肿瘤的一线治疗方法。然而,总生存期和无进展生存期方面的临床结果不如预期.大多数癌症患者对免疫疗法没有反应,原因不同。因此,迫切需要进一步改进癌症免疫疗法.在审查中,我们将讨论正在测试或已经在临床中的各种形式的癌症免疫疗法。此外,我们还强调了改进此类疗法的未来方向。
    The use of cancer immunotherapies is not novel but has been used over the decades in the clinic. Only recently have we found the true potential of stimulating an anti-tumor response after the breakthrough of checkpoint inhibitors. Cancer immunotherapies have become the first line treatment for many malignancies at various stages. Nevertheless, the clinical results in terms of overall survival and progression free survival were not as anticipated. Majority of cancer patients do not respond to immunotherapies and the reasons differ. Hence, further improvements for cancer immunotherapies are crucially needed. In the review, we will discuss various forms of cancer immunotherapies that are being tested or already in the clinic. Moreover, we also highlight future directions to improve such 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
    溶瘤病毒疗法(OVT)已成为一种有前途的癌症免疫疗法,并且由于其增加肿瘤免疫原性和增强宿主抗肿瘤免疫力的能力而能够增强其他免疫疗法。自然杀伤(NK)细胞是介导抗肿瘤反应的关键细胞成分,但他们在调节OVT治疗效果方面的作用享有盛誉.这篇综述将讨论NK细胞如何影响OVT的利弊,以及如何利用这些知识来开发可以调节NK细胞以改善基于OVT的治疗结果的有效策略。
    Oncolytic virotherapy (OVT) has emerged as a promising cancer immunotherapy, and is capable of potentiating other immunotherapies due to its capacity to increase tumor immunogenicity and to boost host antitumor immunity. Natural killer (NK) cells are a critical cellular component for mediating the antitumor response, but hold a mixed reputation for their role in mediating the therapeutic efficacy of OVT. This review will discuss the pros and cons of how NK cells impact OVT, and how to harness this knowledge for the development of effective strategies that could modulate NK cells to improve OVT-based therapeutic outcomes.
    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
    原发性骨恶性肿瘤,包括骨肉瘤(OS),是罕见的,但具有侵略性。当前的操作系统治疗,包括手术切除和化疗,改善了非转移性病例的生存率,但对于复发或转移性OS仍然无效。溶瘤病毒治疗(OVT)是一个有前途的替代方案,使用天然存在的或遗传修饰的病毒来选择性靶向和裂解癌细胞,并诱导针对剩余OS细胞的强大免疫应答。各种溶瘤病毒(OVs),如腺病毒,单纯疱疹病毒,和麻疹病毒,在临床前OS模型中已经证明了疗效。将OVT与其他疗法相结合,如化疗或免疫疗法,可以进一步改善结果。尽管取得了这些进展,临床前模型可靠性的挑战,安全,delivery,和免疫反应必须解决优化OVT的临床应用。未来的研究应该集中在精炼交付方式上,探索联合治疗,和临床试验,以确保OVT的疗效和OS的安全性。总的来说,OVT代表了一种新的方法,有可能极大地改善OS患者的生存结果。
    Primary bone malignancies, including osteosarcoma (OS), are rare but aggressive. Current OS treatment, involving surgical resection and chemotherapy, has improved survival for non-metastatic cases but remains ineffective for recurrent or metastatic OS. Oncolytic viral therapy (OVT) is a promising alternative, using naturally occurring or genetically modified viruses to selectively target and lyse cancer cells and induce a robust immune response against remaining OS cells. Various oncolytic viruses (OVs), such as adenovirus, herpes simplex virus, and measles virus, have demonstrated efficacy in preclinical OS models. Combining OVT with other therapeutics, such as chemotherapy or immunotherapy, may further improve outcomes. Despite these advances, challenges in reliability of preclinical models, safety, delivery, and immune response must be addressed to optimize OVT for clinical use. Future research should focus on refining delivery methods, exploring combination treatments, and clinical trials to ensure OVT\'s efficacy and safety for OS. Overall, OVT represents a novel approach with the potential to drastically improve survival outcomes for patients with OS.
    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:TG6050被设计为一种改进的溶瘤载体,结合痘苗病毒在肿瘤中选择性复制的固有特性与肿瘤限制性表达的重组免疫效应因子来修饰肿瘤免疫表型。这些特性可能对“冷”肿瘤特别感兴趣,浸润不良或浸润无能T细胞。
    方法:溶瘤痘苗病毒TG6050编码单链人白细胞介素-12(hIL-12)和全长抗细胞毒性T淋巴细胞相关抗原-4(@CTLA-4)单克隆抗体。TG6050的相关属性(复制,细胞病,转基因的表达和功能)在体外进行了广泛的表征。病毒载体的生物分布和药代动力学,@CTLA-4和IL-12,以及抗肿瘤活性(单独或与免疫检查点抑制剂组合)在几个“热”(高浸润)和“冷”(低浸润)同系小鼠肿瘤模型中进行了研究。通过监测全身和肿瘤内免疫反应来破译作用机制,和肿瘤转录组分析。在食蟹猴中评估了TG6050反复静脉给药后的安全性,重点关注循环IL-12的水平。
    结果:TG6050在肿瘤细胞体内和体外增殖与功能性IL-12和@CTLA-4的局部表达相关。这种双重机制在“冷”和“热”肿瘤模型(分别为B16F10,LLC1或EMT6,CT26)中都转化为强大的抗肿瘤活性,当与抗程序性细胞死亡蛋白1结合使用时,该活性被进一步放大。对TG6050治疗后肿瘤微环境(TME)变化的分析显示干扰素-γ增加,CD8+T细胞,和M1/M2巨噬细胞比率,以及调节性T细胞的急剧减少。观察到这些局部修饰同时支持系统性和特异性抗肿瘤适应性免疫应答。在毒理学研究中,TG6050在食蟹猴中没有显示任何可观察到的不良反应。
    结论:TG6050有效地将功能性IL-12和@CTLA-4传递到肿瘤中,导致强烈的抗肿瘤活性。向发炎的TME的转变与全身性抗肿瘤T细胞的增加相关。可靠的临床前数据和有利的获益/风险比为TG6050在转移性非小细胞肺癌中的临床评估铺平了道路(NCT05788926试验正在进行中)。
    BACKGROUND: TG6050 was designed as an improved oncolytic vector, combining the intrinsic properties of vaccinia virus to selectively replicate in tumors with the tumor-restricted expression of recombinant immune effectors to modify the tumor immune phenotype. These properties might be of particular interest for \"cold\" tumors, either poorly infiltrated or infiltrated with anergic T cells.
    METHODS:  TG6050, an oncolytic vaccinia virus encodes single-chain human interleukin-12 (hIL-12) and full-length anti-cytotoxic T-lymphocyte-associated antigen-4 (@CTLA-4) monoclonal antibody. The relevant properties of TG6050 (replication, cytopathy, transgenes expression and functionality) were extensively characterized in vitro. The biodistribution and pharmacokinetics of the viral vector, @CTLA-4 and IL-12, as well as antitumoral activities (alone or combined with immune checkpoint inhibitors) were investigated in several \"hot\" (highly infiltrated) and \"cold\" (poorly infiltrated) syngeneic murine tumor models. The mechanism of action was deciphered by monitoring both systemic and intratumoral immune responses, and by tumor transcriptome analysis. The safety of TG6050 after repeated intravenous administrations was evaluated in cynomolgus monkeys, with a focus on the level of circulating IL-12.
    RESULTS: Multiplication and propagation of TG6050 in tumor cells in vitro and in vivo were associated with local expression of functional IL-12 and @CTLA-4. This dual mechanism translated into a strong antitumoral activity in both \"cold\" and \"hot\" tumor models (B16F10, LLC1 or EMT6, CT26, respectively) that was further amplified when combined with anti-programmed cell death protein-1. Analysis of changes in the tumor microenvironment (TME) after treatment with TG6050 showed increases in interferon-gamma, of CD8+T cells, and of M1/M2 macrophages ratio, as well as a drastic decrease of regulatory T cells. These local modifications were observed alongside bolstering a systemic and specific antitumor adaptive immune response. In toxicology studies, TG6050 did not display any observable adverse effects in cynomolgus monkeys.
    CONCLUSIONS: TG6050 effectively delivers functional IL-12 and @CTLA-4 into the tumor, resulting in strong antitumor activity. The shift towards an inflamed TME correlated with a boost in systemic antitumor T cells. The solid preclinical data and favorable benefit/risk ratio paved the way for the clinical evaluation of TG6050 in metastatic non-small cell lung cancer (NCT05788926 trial in progress).
    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:在过去的十年中,癌症免疫疗法彻底改变了黑色素瘤的治疗方法;然而,不同患者人群的反应各不相同。最近,在接受免疫检查点抑制剂治疗的黑色素瘤患者中,基线肿瘤大小已被确定为总生存期的独立预后因素.MG1是一种具有广泛肿瘤嗜性的新型溶瘤药物,最近已进入早期临床试验。这项研究的目的是表征MG1治疗后人类和小鼠黑色素瘤模型中的T细胞反应,并确定在两种不同肿瘤负荷下肿瘤免疫微环境(TIME)的特征是否会影响溶瘤病毒疗法的功效。
    方法:进行人三维体外引发测定以测量MG1感染后的抗肿瘤和抗病毒T细胞应答。T细胞受体(TCR)测序,T2杀伤试验,和肽召回分析用于评估TCR库的演变,并测量特定的T细胞反应,分别。在体内,皮下4434黑色素瘤使用RNA测序进行表征,免疫组织化学,和流式细胞术。肿瘤内MG1的有效性在推进4434肿瘤中进行了评估,并通过脾细胞回忆测定法测量了抗肿瘤和抗病毒T细胞的产生。最后,在4434例晚期肿瘤中研究了MG1和程序性细胞死亡蛋白-1抗体(αPD-1)的联合治疗。
    结果:MG1有效地支持功能性细胞毒性T细胞(CTL)针对肿瘤相关抗原以及病毒衍生肽的启动,如使用肽召回和T2杀伤分析评估,分别。TCR测序显示,与对照相比,MG1引发的CTL包含更大的相似CDR3氨基酸序列簇。MG1的体内测试表明,MG1单一疗法在治疗早期疾病方面非常有效,导致90%的治愈;然而,MG1的疗效随着疾病负担(局部肿瘤大小)的增加而降低,在更晚期的疾病中,需要添加αPD-1来克服耐药性。差异基因表达谱显示,肿瘤负荷增加与免疫学上较冷的TIME相关。此外,与较小的肿瘤相比,在进展肿瘤中的TCR信号分析显示了TCR参与的不同动态,特别是CD4+细胞的抗原识别发生了变化,从常规子集到监管子集。
    结论:需要向MG1中添加αPD-1以克服免疫学上“较冷”更晚期黑色素瘤的病毒治疗耐药性,强调肿瘤负担对不同类型免疫治疗的重要性。
    BACKGROUND: Over the past decade, cancer immunotherapies have revolutionized the treatment of melanoma; however, responses vary across patient populations. Recently, baseline tumor size has been identified as an independent prognostic factor for overall survival in patients with melanoma receiving immune checkpoint inhibitors. MG1 is a novel oncolytic agent with broad tumor tropism that has recently entered early-phase clinical trials. The aim of this study was to characterize T-cell responses in human and mouse melanoma models following MG1 treatment and to establish if features of the tumor immune microenvironment (TIME) at two distinct tumor burdens would impact the efficacy of oncolytic virotherapy.
    METHODS: Human three-dimensional in vitro priming assays were performed to measure antitumor and antiviral T-cell responses following MG1 infection. T-cell receptor (TCR) sequencing, T2 killing assay, and peptide recall assays were used to assess the evolution of the TCR repertoire, and measure specific T-cell responses, respectively. In vivo, subcutaneous 4434 melanomas were characterized using RNA sequencing, immunohistochemistry, and flow cytometry. The effectiveness of intratumoral MG1 was assessed in advancing 4434 tumors and the generation of antitumor and antiviral T cells measured by splenocyte recall assays. Finally, combination MG1 and programmed cell death protein-1 antibody (αPD-1) therapy was investigated in advanced 4434 tumors.
    RESULTS: MG1 effectively supported priming of functional cytotoxic T cells (CTLs) against tumor-associated antigens as well as virus-derived peptides, as assessed using peptide recall and T2 killing assays, respectively. TCR sequencing revealed that MG1-primed CTL comprised larger clusters of similar CDR3 amino acid sequences compared with controls. In vivo testing of MG1 demonstrated that MG1 monotherapy was highly effective at treating early disease, resulting in 90% cures; however, the efficacy of MG1 reduced as the disease burden (local tumor size) increased, and the addition of αPD-1 was required to overcome resistance in more advanced disease. Differential gene expression profiles revealed that increased tumor burden was associated with an immunologically colder TIME. Furthermore, analysis of TCR signaling in advancing tumors demonstrated a different dynamic of TCR engagement compared with smaller tumors, in particular a shift in antigen recognition by CD4+ cells, from conventional to regulatory subsets.
    CONCLUSIONS: Addition of αPD-1 to MG1 is required to overcome viral therapy resistance in immunologically \'colder\' more advanced melanoma, highlighting the importance of tumor burden to different types of immunotherapy.
    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
    溶瘤病毒疗法有望为癌症提供新的治疗策略。柏油凝集素(AVL)是一种Ca2依赖性凝集素受体,含有C型凝集素的保守结构域和疏水性N末端区域,能与鸟巢糖蛋白和D-半乳糖结合。我们先前的研究表明,携带AVL基因的溶瘤痘苗病毒(oncoVV)在体内外具有显着的复制和抗肿瘤作用。在这项研究中,我们发现oncoVV-AVL可能重新编程肝癌细胞的代谢以促进ROS,和升高的ROS随后促进病毒复制并诱导细胞凋亡。本研究将为肿瘤VV-AVL在肝癌中的应用提供新的理论依据。
    Oncolytic virotherapy is expected to provide a new treatment strategy for cancer. Aphrocallistes vastus lectin (AVL) is a Ca2+-dependent lectin receptor containing the conserved domain of C-type lectin and the hydrophobic N-terminal region, which can bind to the bird\'s nest glycoprotein and D-galactose. Our previous studies suggested that the oncolytic vaccinia virus (oncoVV) armed with the AVL gene exerted remarkable replication and antitumor effects in vitro and in vivo. In this study, we found that oncoVV-AVL may reprogram the metabolism of hepatocellular carcinoma cells to promote ROS, and elevated ROS subsequently promoted viral replication and induced apoptosis. This study will provide a new theoretical basis for the application of oncoVV-AVL in liver cancer.
    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
    恶性周围神经鞘瘤(MPNST)由于手术切除后的高复发率和对传统化疗的无效反应而提出了重大的治疗挑战。另一种治疗策略是溶瘤病毒免疫疗法,它可以引发持久和系统性的抗肿瘤免疫反应,并被食品和药物管理局(FDA)批准用于治疗黑色素瘤。不幸的是,只有一部分患者完全有反应,强调需要解决阻碍病毒免疫疗法有效性的障碍。
    在这里,我们研究了靶向MPNST免疫抑制微环境的关键成分以增强病毒免疫疗法在三种小鼠模型中的抗肿瘤功效的治疗效用,其中一个显示出比其他更多的免疫原性特征。
    用帕西达替尼进行骨髓调节治疗,CSF1R酪氨酸激酶的小分子抑制剂,在高免疫原性模型中,溶瘤性单纯疱疹病毒T-VEC的中位生存时间增加最显著。此外,用骨髓调节疗法靶向骨髓细胞,caspase-8依赖性凋亡的小分子激活剂,在免疫原性较低的MPNST模型中增加了T-VEC的生存益处。然而,未观察到肿瘤消退或缩小.耗竭实验证实,增强的存活益处依赖于T细胞应答。此外,联合病毒免疫疗法后的流式细胞术分析显示,肿瘤微环境中M2巨噬细胞和髓源性抑制细胞减少,肿瘤特异性gp70+CD8T细胞增加.
    总之,我们的研究结果为利用针对MPNST的骨髓细胞靶向的病毒免疫疗法的潜力提供了令人信服的证据,值得进一步研究.
    UNASSIGNED: Malignant peripheral nerve sheath tumors (MPNST) pose a significant therapeutic challenge due to high recurrence rates after surgical resection and a largely ineffective response to traditional chemotherapy. An alternative treatment strategy is oncolytic viroimmunotherapy, which can elicit a durable and systemic antitumor immune response and is Food and Drug Administration (FDA)-approved for the treatment of melanoma. Unfortunately, only a subset of patients responds completely, underscoring the need to address barriers hindering viroimmunotherapy effectiveness.
    UNASSIGNED: Here we investigated the therapeutic utility of targeting key components of the MPNST immunosuppressive microenvironment to enhance viroimmunotherapy\'s antitumor efficacy in three murine models, one of which showed more immunogenic characteristics than the others.
    UNASSIGNED: Myelomodulatory therapy with pexidartinib, a small molecule inhibitor of CSF1R tyrosine kinase, and the oncolytic herpes simplex virus T-VEC exhibited the most significant increase in median survival time in the highly immunogenic model. Additionally, targeting myeloid cells with the myelomodulatory therapy trabectedin, a small molecule activator of caspase-8 dependent apoptosis, augmented the survival benefit of T-VEC in a less immunogenic MPNST model. However, tumor regressions or shrinkages were not observed. Depletion experiments confirmed that the enhanced survival benefit relied on a T cell response. Furthermore, flow cytometry analysis following combination viroimmunotherapy revealed decreased M2 macrophages and myeloid-derived suppressor cells and increased tumor-specific gp70+ CD8 T cells within the tumor microenvironment.
    UNASSIGNED: In summary, our findings provide compelling evidence for the potential to leverage viroimmunotherapy with myeloid cell targeting against MPNST and warrant further investigation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号