oncolytic viruses

溶瘤病毒
  • 文章类型: Journal Article
    小儿脑肿瘤是儿童中最常见的实体瘤。即使到目前为止,随着多模态治疗管理的进步,在某些类型的肿瘤中,生存结果仍然令人沮丧,如小儿型弥漫性高级别胶质瘤或中枢神经系统(CNS)胚胎性肿瘤。未能理解复杂的分子异质性以及难以捉摸的肿瘤和微环境相互作用继续破坏治疗功效。在儿科神经肿瘤学中,开发一种改善这些致命肿瘤生存率的策略仍未实现。溶瘤病毒(OVs)正在成为一种可行的病毒,安全,和有希望的治疗脑肿瘤。病毒疗法的新范式意味着遵循直接的细胞病变效应,在某些情况下,通过抗肿瘤免疫反应来部分或完全消除肿瘤块。OVs单独或与其他治疗方式组合已主要用于成人神经肿瘤学。最近,儿科脑肿瘤模型中令人鼓舞的临床前研究激增,导致OVs的临床转化,并在这些肿瘤中取得了令人鼓舞的结果。在这次审查中,我们总结了在小儿脑肿瘤的临床前和临床研究中测试的不同病毒疗法,我们讨论了改善这些肿瘤对这种治疗的反应所必需的局限性和未来的途径。
    Pediatric brain tumors are the most common solid tumors in children. Even to date, with the advances in multimodality therapeutic management, survival outcomes remain dismal in some types of tumors, such as pediatric-type diffuse high-grade gliomas or central nervous system (CNS) embryonal tumors. Failure to understand the complex molecular heterogeneity and the elusive tumor and microenvironment interplay continues to undermine therapeutic efficacy. Developing a strategy that would improve survival for these fatal tumors remains unmet in pediatric neuro-oncology. Oncolytic viruses (OVs) are emerging as a feasible, safe, and promising therapy for brain tumors. The new paradigm in virotherapy implies that the direct cytopathic effect is followed, under certain circumstances, by an antitumor immune response responsible for the partial or complete debulking of the tumor mass. OVs alone or combined with other therapeutic modalities have been primarily used in adult neuro-oncology. A surge in encouraging preclinical studies in pediatric brain tumor models recently led to the clinical translation of OVs with encouraging results in these tumors. In this review, we summarize the different virotherapy tested in preclinical and clinical studies in pediatric brain tumors, and we discuss the limitations and future avenues necessary to improve the response of these tumors to this type of therapy.
    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,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
    40多年前在活化T细胞的上清液中发现,白细胞介素-2(IL-2)是一种有效的多效性细胞因子,参与免疫反应的调节。它是效应T细胞扩增和分化以及调节性T细胞诱导的外周耐受性所必需的。大剂量IL-2治疗是FDA批准的第一个针对肾细胞癌和黑色素瘤的免疫疗法。实现单一主体完整和持久的响应,虽然只有一小部分患者。野生型IL-2的治疗潜力在临床上受到其短半衰期和严重血管毒性的限制。此外,调节性T细胞的活化和效应性T细胞对IL-2的终末分化构成了额外的限制。为了克服IL-2的毒性,以实现其对患者的全部潜力,正在开发几种新的工程策略,基于IL-2的癌症免疫治疗已成为临床和实验研究的新兴领域。此外,在临床前肿瘤模型中,IL-2与PD-1/L1通路阻断的联合治疗显示出显著改善的抗肿瘤疗效.本文综述了IL-2及其受体的生物学特性,以及其在癌症患者中的疗效和治疗限制性毒性。我们还探索了旨在开发新的和更安全的IL-2疗法的努力,以利用该细胞因子的全部治疗潜力。
    Discovered over 4 decades ago in the supernatants of activated T cells, interleukin-2 (IL-2) is a potent pleiotropic cytokine involved in the regulation of immune responses. It is required for effector T cell expansion and differentiation as well as for peripheral tolerance induced by regulatory T cells. High-dose IL-2 treatment was the first FDA-approved immunotherapy for renal cell carcinoma and melanoma, achieving single agent complete and durable responses, albeit only in a small proportion of patients. The therapeutic potential of wild type IL-2 is clinically limited by its short half-life and severe vascular toxicity. Moreover, the activation of regulatory T cells and the terminal differentiation of effector T cells on IL-2 pose additional restrictions. To overcome the toxicity of IL-2 in order to realize its full potential for patients, several novel engineering strategies are being developed and IL-2 based immunotherapy for cancer has emerged as a burgeoning field of clinical and experimental research. In addition, combination of IL-2 with PD-1/L1 pathway blockade shows vastly improved anti-tumor efficacy over either monotherapy in preclinical tumor models. In this review we discuss the biological characteristics of IL-2 and its receptors, as well as its efficacy and treatment limiting toxicities in cancer patients. We also explore the efforts aimed at developing novel and safer IL-2 therapies to harness the full therapeutic potential of this cytokine.
    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
    呼肠孤病毒(Reo)在特异性杀死肿瘤细胞方面显示出有希望的潜力,并为卵巢癌(OC)治疗提供了新的可能性。然而,OC患者腹水中的中和抗体极大地限制了Reo的进一步应用。在这项研究中,我们使用阳离子脂质体(Lipo)来递送Reo,显着增强其进入OC细胞的能力及其在腹水条件下杀死这些细胞的有效性。用MβCD抑制剂预处理显著降低Reo介导的肿瘤细胞死亡,表明Lipo主要通过小窝蛋白介导的内吞作用使Reo的细胞摄取成为可能。我们的结果表明,Lipo有效地促进Reo进入细胞质并触发细胞凋亡。上述发现为克服中和抗体在Reo临床应用中的障碍提供了新的策略。
    Reovirus (Reo) has shown promising potential in specifically killing tumor cells, and offering new possibilities for ovarian cancer (OC) treatment. However, neutralizing antibodies in the ascites from OC patients greatly limit the further application of Reo. In this study, we employed cationic liposomes (Lipo) to deliver Reo, significantly enhancing its ability to enter OC cells and its effectiveness in killing these cells under ascitic conditions. Pre-treatment with the MβCD inhibitor notably decreased Reo-mediated tumor cell death, indicating that Lipo primarily enables Reo\'s cellular uptake through caveolin-mediated endocytosis. Our results demonstrate that Lipo effectively facilitates the entry of Reo into the cytoplasm and triggers cell apoptosis. The above findings provide a new strategy to overcome the obstacle of neutralizing antibodies in the clinical application of Reo.
    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
    探讨溶瘤禽呼肠孤病毒(ARV)p17蛋白是否介导细胞迁移和侵袭足形成,我们应用了几种分子生物学方法来研究相关的细胞因子和信号通路。我们发现ARVp17激活p53/磷酸酶和张力蛋白同源物(PTEN)通路,抑制粘着斑激酶(FAK)/Src信号和下游信号分子,从而抑制小鼠黑色素瘤细胞系(B16-F10)中的细胞迁移和侵袭性足病的形成。重要的是,在用突变体PTENC124A转染的细胞中,p17诱导的invadopodia形成可以逆转。发现p17蛋白显着降低酪氨酸激酶底物5(TKs5)的表达水平,Rab40b,酪氨酸激酶衔接蛋白1(NCK1)的非催化区,和基质金属蛋白酶(MMP9),表明TKs5和Rab40b被p17转录下调。此外,我们发现p17抑制了TKs5/NCK1复合物的形成。B16-F10癌细胞中TKs5和Rab40b的共表达逆转了p17调节的对侵袭性足病形成的抑制。这项工作提供了通过激活p53/PTEN通路来抑制p17调节的invadopodia形成的新见解,抑制FAK/Src通路,并抑制TKs5/NCK1复合物的形成。
    To explore whether the p17 protein of oncolytic avian reovirus (ARV) mediates cell migration and invadopodia formation, we applied several molecular biological approaches for studying the involved cellular factors and signal pathways. We found that ARV p17 activates the p53/phosphatase and tensin homolog (PTEN) pathway to suppress the focal adhesion kinase (FAK)/Src signaling and downstream signal molecules, thus inhibiting cell migration and the formation of invadopodia in murine melanoma cancer cell line (B16-F10). Importantly, p17-induced formation of invadopodia could be reversed in cells transfected with the mutant PTENC124A. p17 protein was found to significantly reduce the expression levels of tyrosine kinase substrate 5 (TKs5), Rab40b, non-catalytic region of tyrosine kinase adaptor protein 1 (NCK1), and matrix metalloproteinases (MMP9), suggesting that TKs5 and Rab40b were transcriptionally downregulated by p17. Furthermore, we found that p17 suppresses the formation of the TKs5/NCK1 complex. Coexpression of TKs5 and Rab40b in B16-F10 cancer cells reversed p17-modulated suppression of the formation of invadopodia. This work provides new insights into p17-modulated suppression of invadopodia formation by activating the p53/PTEN pathway, suppressing the FAK/Src pathway, and inhibiting the formation of the TKs5/NCK1 complex.
    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
    由于引入了免疫检查点抑制剂(ICI),最近皮肤癌的预后得到了极大改善。然而,许多晚期皮肤癌患者在ICI治疗期间仍然经历免疫疗法抵抗和疾病进展,因此需要解决这一治疗差距的新疗法。近年来,Talimogenelaherparepvec(T-VEC)已成为皮肤癌患者的可行治疗选择。在临床前研究中,T-VEC在注射性病变中表现出直接的抗肿瘤作用以及在非注射性病变中表现出全身免疫介导的作用。与ICI治疗联合使用时可能会带来额外的益处。根据OPTIM试验的有希望的结果,美国食品和药物管理局(FDA)批准T-VEC作为单一药物治疗晚期黑色素瘤.然而,MASTERKEY-265试验表明,在pembrolizumab中加入T-VEC并不能为黑色素瘤患者带来额外的临床获益.然而,T-VEC的有希望的疗效和FDA的批准帮助溶瘤病毒(OVs)在癌症治疗中获得广泛关注,并且已经进行了广泛的研究来评估OVs在其他肿瘤如肉瘤和乳腺癌中的使用。这里,我们回顾了2022年至2024年的临床结果,这些结果调查了OVs作为单药或与其他疗法联合治疗皮肤恶性肿瘤的有效性和安全性.此外,我们描述了目前OV利用的局限性,并概述了未来的方向,以提高接受OV治疗的皮肤恶性肿瘤患者的临床结局.
    Skin cancer prognosis has greatly improved recently due to the introduction of immune checkpoint inhibitors (ICIs). However, many patients with advanced skin cancer still experience immunotherapy resistance and disease progression during ICI treatment, thus calling for novel therapeutics which address this treatment gap. Talimogene laherparepvec (T-VEC) has gained popularity in recent years as a viable treatment option for patients with skin cancer. In preclinical studies, T-VEC demonstrated both a direct anti-tumor effect in injected lesions as well as a systemic immune-mediated effect in non-injected lesions, which could pose additional benefits when combined with ICI therapy. Following promising results from the OPTiM trial, the Food and Drug Administration (FDA) approved the usage of T-VEC as a single agent in advanced melanoma. However, the MASTERKEY-265 trial demonstrated that adding T-VEC to pembrolizumab did not offer additional clinical benefit in patients with melanoma. Nevertheless, the promising efficacy of T-VEC and its approval by the FDA helped oncolytic viruses (OVs) gain wide attention in cancer therapy, and extensive research has been undertaken to evaluate the usage of OVs in other tumors such as sarcomas and breast cancers. Here, we provide a review of clinical results from 2022 to 2024 that investigate the efficacy and safety of OVs as a monotherapy or in combination with other therapies in skin malignancies. Furthermore, we delineate the current limitations in OV utilization and outline future directions to enhance clinical outcomes for patients with skin malignancies receiving OV-based 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
    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)

公众号