T-VEC

T - VEC
  • 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
    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)

  • 文章类型: Journal Article
    [这修正了文章DOI:10.3389/fimmu.2024.1384623。].
    [This corrects the article DOI: 10.3389/fimmu.2024.1384623.].
    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
    睾丸核蛋白癌(NC)是一种高度侵袭性的肿瘤疾病。鉴于目前的治疗方案只能提供6个月的中位生存期,这种类型的肿瘤很可能需要扩展的多模式治疗方法来改善预后.在之前的病例报告中,我们可以证明溶瘤单纯疱疹病毒(T-VEC)在NC患者中具有功能。为了确定T-VEC的进一步组合合作伙伴,我们已经研究了T-VEC和5种不同的小分子抑制剂(SMI)在人NC细胞系中单独和组合的抗肿瘤作用。发现与各自的单一疗法相比,双重组合导致更高的肿瘤细胞减少率,如通过活力测定和实时肿瘤细胞生长监测所证明的。有趣的是,我们发现T-VEC与SMI的组合导致c-Myc表达的更强且更早的减少,NC细胞增殖的主要驱动因素,与T-VEC单一疗法相比。这些结果表明,使用溶瘤病毒和SMI的组合疗法具有控制NC癌症的高度攻击行为的巨大潜力,并且可能会在不久的将来为创新的多模式临床研究铺平道路。
    NUT (nuclear-protein-in-testis) carcinoma (NC) is a highly aggressive tumor disease. Given that current treatment regimens offer a median survival of six months only, it is likely that this type of tumor requires an extended multimodal treatment approach to improve prognosis. In an earlier case report, we could show that an oncolytic herpes simplex virus (T-VEC) is functional in NC patients. To identify further combination partners for T-VEC, we have investigated the anti-tumoral effects of T-VEC and five different small molecule inhibitors (SMIs) alone and in combination in human NC cell lines. Dual combinations were found to result in higher rates of tumor cell reductions when compared to the respective monotherapy as demonstrated by viability assays and real-time tumor cell growth monitoring. Interestingly, we found that the combination of T-VEC with SMIs resulted in both stronger and earlier reductions in the expression of c-Myc, a main driver of NC cell proliferation, when compared to T-VEC monotherapy. These results indicate the great potential of combinatorial therapies using oncolytic viruses and SMIs to control the highly aggressive behavior of NC cancers and probably will pave the way for innovative multimodal clinical studies in the near 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: Case Reports
    背景:粘膜黑色素瘤,一种与头颈部常见的皮肤黑色素瘤不同的侵袭性恶性肿瘤,占所有恶性黑色素瘤的<1%。粘膜黑色素瘤的发病机制尚不清楚。在皮肤黑色素瘤中常见的靶向突变,比如在BRAF和NRAS基因中,粘膜黑色素瘤的发病率较低。粘膜黑素瘤携带与皮肤黑素瘤不同的突变模式。切缘阴性的手术是粘膜黑色素瘤的一线治疗,和系统治疗并不明确。Talimoenelaherparepvec,溶瘤病毒免疫疗法,是美国食品和药物管理局批准用于治疗晚期恶性皮肤黑色素瘤,具有局部治疗益处。粘膜黑色素瘤最初被排除在talimogenelaherparepvec的初始III期临床试验之外。
    方法:我们介绍了一名40多岁的白人女性患者,既往有系统性红斑狼疮病史,硬皮病,和雌激素受体阳性浸润性导管癌。双侧乳房切除术后,患者被发现患有BRAF阴性的硬腭粘膜黑色素瘤伴软腭跳跃病变.由于存在跳跃粘膜病变以及预期的缺损和需要游离皮瓣重建手术,考虑非手术治疗.病人被转诊到肿瘤内科,根据患者的复杂病史和免疫治疗的风险,可能使她先前的自身免疫性疾病恶化,选择了局部talimogenelaherparepvec注射作为她的粘膜病变的主要治疗方法。尽管talimogenelaherparepvec被批准用于皮肤黑色素瘤的治疗,关于在粘膜黑素瘤中使用talimogenelaherparepvec的可用数据有限。
    结论:患者在原发病灶以及局部注射后的跳跃病灶均有完全的局部肿瘤反应。她没有副作用,在治疗期间保持了高质量的生活。
    BACKGROUND: Mucosal melanoma, an aggressive type of malignancy different from the cutaneous melanomas commonly seen in the head and neck region, represents < 1% of all malignant melanomas. The pathogenesis of mucosal melanoma is unknown. Targetable mutations commonly seen in cutaneous melanoma, such as in the BRAF and NRAS genes, have a lower incidence in mucosal melanoma. Mucosal melanoma carries a distinct mutational pattern from cutaneous melanoma. Surgery with negative margins is the first-line treatment for mucosal melanoma, and systemic therapy is not well defined. Talimogene laherparepvec, an oncolytic viral immunotherapy, is United States Food and Drug Administration approved for the treatment of advanced malignant cutaneous melanoma, with local therapeutic benefits. Mucosal melanoma was initially excluded from talimogene laherparepvec\'s initial phase III clinical trial.
    METHODS: We present the case of a white female patient in her 40s with past medical history of systemic lupus erythematous, scleroderma, and estrogen-receptor-positive invasive ductal breast carcinoma. Following a bilateral mastectomy, the patient was found to have BRAF-negative mucosal melanoma of her hard palate with a soft palate skip lesion. Owing to the presence of a skip mucosal lesion as well as the anticipated defect and need for free-flap reconstructive surgery, nonsurgical management was considered. The patient was referred to medical oncology, where-based on the patient\'s complicated medical history and the risk of immunotherapy possibly worsening her prior autoimmune diseases-local talimogene laherparepvec injections were chosen as the primary therapy for her mucosal lesions. Though talimogene laherparepvec is approved for the treatment of cutaneous melanoma, there are limited data available on the use of talimogene laherparepvec in mucosal melanomas.
    CONCLUSIONS: The patient had a complete local tumor response at both the primary lesion as well as the skip lesion with the local injections. She had no side effects and maintained a high quality of life during 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:溶瘤病毒疗法(OVT)是一种有前途的抗肿瘤方式,它利用溶瘤病毒(OVs)优先攻击癌症而不是正常组织。随着对病毒和肿瘤细胞特性的了解,许多创新的OV被设计用来征服癌症,例如塔利莫根拉赫帕雷普维奇(T-VEC)和塔萨纳托雷夫(DNX-2401)。然而,必须进一步优化和平衡治疗安全性和有效性,以确保临床上的OVT具有出色的安全性和有效性,合理的联合治疗策略也是值得探讨的重要挑战。
    方法:在这里,我们对OVT的发展历史和现状进行了严格的回顾,强调提高安全性和有效性的机制。我们认为溶瘤病毒疗法已经发展成为第四代肿瘤免疫疗法。特别是,通过设计表达双特异性T细胞激活剂(BiTA)的OV来激发T细胞是一种用一块石头杀死两只鸟的有希望的策略。OVs和免疫细胞的治疗策略的惊人组合赋予了管理癌症的巨大潜力。此外,最近讨论了有吸引力的临床前OVT,并对临床试验中的OVT进行了系统评价。
    结论:OVs,正在进入临床试验,即将成为临床抗肿瘤药物的前沿。
    Oncolytic virotherapy (OVT) is a promising anti-tumor modality that utilizes oncolytic viruses (OVs) to preferentially attack cancers rather than normal tissues. With the understanding particularly in the characteristics of viruses and tumor cells, numerous innovative OVs have been engineered to conquer cancers, such as Talimogene Laherparepvec (T-VEC) and tasadenoturev (DNX-2401). However, the therapeutic safety and efficacy must be further optimized and balanced to ensure the superior safe and efficient OVT in clinics, and reasonable combination therapy strategies are also important challenges worthy to be explored.
    Here we provided a critical review of the development history and status of OVT, emphasizing the mechanisms of enhancing both safety and efficacy. We propose that oncolytic virotherapy has evolved into the fourth generation as tumor immunotherapy. Particularly, to arouse T cells by designing OVs expressing bi-specific T cell activator (BiTA) is a promising strategy of killing two birds with one stone. Amazing combination of therapeutic strategies of OVs and immune cells confers immense potential for managing cancers. Moreover, the attractive preclinical OVT addressed recently, and the OVT in clinical trials were systematically reviewed.
    OVs, which are advancing into clinical trials, are being envisioned as the frontier clinical anti-tumor agents coming soon.
    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
    角质细胞癌(KCs),特别是皮肤鳞状细胞癌和基底细胞癌,可以对局部做出反应,病灶内,或全身免疫疗法,但皮肤不良事件(CAE)可能发生。了解这些风险,早期识别这些CAE,和有效的治疗可以使患者继续他们的抗癌免疫疗法没有剂量的影响。KCs后免疫检查点抑制剂相关的CAE可以有多种临床表现,观察到的具体类型包括银屑病和大疱性类天疱疮。皮肤毒性可能需要活检来确认诊断,尤其是对局部或口服类固醇无反应的患者,因为生物药物的选择取决于准确的诊断。来自免疫检查点抑制剂的不同类型的CAE与各种原发性癌症类型的不同肿瘤学结果相关。这仍有待KC患者确定。KC患者免疫检查点抑制剂后的CAE表征和管理是一个快速增长的领域,需要具体和前瞻性研究。
    Keratinocytic cancers (KCs), specifically cutaneous squamous cell and basal cell carcinomas, can respond to topical, intralesional, or systemic immunotherapies, but cutaneous adverse events (CAEs) may occur. Understanding these risks, early recognition of these CAEs, and effective treatment may enable patients to continue their anticancer immunotherapies without dose impact. Immune checkpoint inhibitor-related CAEs after KCs can have multiple clinical presentations, with specific observed types including psoriasis and bullous pemphigoid. Cutaneous toxicities can require biopsies to confirm the diagnosis, especially in patients who are not responsive to topical or oral steroids, since the selection of biologic drugs depends on accurate diagnosis. Different types of CAEs from immune checkpoint inhibitors have been associated with different oncologic outcomes in various primary cancer types, and this remains to be determined for KC patients. CAE characterization and management after immune checkpoint inhibitors in KC patients is a rapidly growing field that needs specific and prospective studies.
    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
    暂无摘要。
    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
    在晚期和高风险黑色素瘤的治疗中使用免疫疗法已导致结果的显着改善。尽管黑色素瘤的发病率持续上升,晚期无法手术的IV期疾病患者的中位生存期从约6个月提高到近6年.最近对肿瘤微环境及其与免疫系统相互作用的理解导致了新型免疫疗法的爆炸性发展。自1990年代批准治疗性细胞因子白介素-2和干扰素α-2以来,新型免疫检查点抑制剂(ICIs)的开发,溶瘤病毒治疗,和肿瘤微环境的调节剂已经让位于黑色素瘤治疗的新时代。针对程序性细胞死亡蛋白1受体(PD-1)及其配体(PDL-1)的单克隆抗体,细胞毒性T淋巴细胞相关蛋白4(CTLA-4),和淋巴细胞激活基因3(LAG-3)提供了适应性免疫系统的强大激活,恢复导致宿主肿瘤识别和破坏的免疫监视。其他多种免疫调节疗法正在研究中,以克服对ICI治疗的耐药性。包括toll样受体-9(TLR-9)和7/8(TLR-7/8)激动剂,干扰素基因刺激因子(STING)激动剂,和粪便微生物移植。在这次审查中,我们关注免疫治疗治疗黑色素瘤的最新进展,并对目前正在研究的新疗法进行了更新.
    The use of immunotherapy in the treatment of advanced and high-risk melanoma has led to a striking improvement in outcomes. Although the incidence of melanoma has continued to rise, median survival has improved from approximately 6 months to nearly 6 years for patients with advanced inoperable stage IV disease. Recent understanding of the tumor microenvironment and its interplay with the immune system has led to the explosive development of novel immunotherapy treatments. Since the approval of the therapeutic cytokines interleukin-2 and interferon alfa-2 in the 1990s, the development of novel immune checkpoint inhibitors (ICIs), oncolytic virus therapy, and modulators of the tumor microenvironment have given way to a new era in melanoma treatment. Monoclonal antibodies directed at programmed cell death protein 1 receptor (PD-1) and its ligand (PDL-1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), and lymphocyte-activation gene 3 (LAG-3) have provided robust activation of the adaptive immune system, restoring immune surveillance leading to host tumor recognition and destruction. Multiple other immunomodulatory therapeutics are under investigation to overcome resistance to ICI therapy, including the toll-like receptor-9 (TLR-9) and 7/8 (TLR-7/8) agonists, stimulator of interferon genes (STING) agonists, and fecal microbiota transplantation. In this review, we focus on the recent advances in immunotherapy for the treatment of melanoma and provide an update on novel therapies currently under investigation.
    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
    在过去的十年中,黑色素瘤治疗的前景经历了一场戏剧性的革命。溶瘤病毒(OVs)的使用代表了一种新颖的治疗方法,可以选择性地感染和裂解肿瘤细胞并诱导局部和全身抗肿瘤免疫反应。作为食品和药物管理局(FDA)批准用于黑色素瘤治疗的第一个OV,talimogenelaherparepvec(T-VEC),转基因单纯疱疹病毒(HSV),在晚期黑色素瘤的治疗中显示出有希望的治疗效果,作为单一疗法或与其他免疫疗法组合,例如免疫检查点抑制剂(ICIs)。经过验证的疗效,T-VEC已在临床试验环境中针对多种其他癌症类型进行了评估。在这篇文章中,我们将对OVs以及T-VEC在黑色素瘤单一治疗和联合治疗中的应用进行综述.此外,我们将综述T-VEC在其他类型皮肤癌中的应用进展。此外,我们将简要描述我们在希望之城接受T-VEC治疗的经验,旨在为扩展其未来应用提供更多见解。
    The landscape of melanoma treatment has undergone a dramatic revolution in the past decade. The use of oncolytic viruses (OVs) represents a novel therapeutic approach that can selectively infect and lyse tumor cells and induce local and systemic antitumor immune responses. As the first OV approved by the Food and Drug Administration (FDA) for melanoma treatment, talimogene laherparepvec (T-VEC), a genetically modified herpes simplex virus (HSV), has shown promising therapeutic effects in the treatment of advanced melanoma, both as a monotherapy or in combination with other immunotherapies, such as the immune checkpoint inhibitors (ICIs). With proven efficacy, T-VEC has been evaluated against a variety of other cancer types in a clinical trial setting. In this article, we will provide a review on OVs and the application of T-VEC in melanoma monotherapy and combination therapy. In addition, we will review the recent progress of T-VEC application in other cutaneous cancer types. Moreover, we will briefly describe our experience of T-VEC therapy at City of Hope, aiming to provide more insight for expanding its future application.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号