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: Editorial
    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: Case Reports
    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: Case Reports
    NRAS基因突变是恶性黑色素瘤的常见改变。然而,除了免疫检查点抑制外,对于NRAS突变的黑色素瘤患者,没有批准的特定治疗方案.由于临床前数据表明MEK抑制剂(MEKi)和溶瘤病毒talimogenelaherparepvec(T-VEC)的协同作用,我们已经用这种组合治疗了三名黑色素瘤患者。所有三名患者都患有复发性皮肤和皮下运输转移。治疗后,一名患者(病例1)表现出局部转移完全消退,并且至今仍无进展,差不多三年了.第二例患者(病例2)显示出痛苦的臀部卫星转移的部分消退,但因脑转移而死亡。第三例患者(病例3)表现出持续7个月的局部转移反应。联合治疗对每种单一药物已知的常见不良事件具有良好的耐受性。本报告是第一个病例系列,介绍T-VEC和MEKi联合治疗的临床益处。我们建议T-VEC和MEKi的组合作为NRAS突变患者的非标记治疗选择。特别是在途中复发或卫星转移。
    Mutations in the NRAS gene are common alterations in malignant melanoma. However, there are no specific treatment options approved for NRAS-mutated melanoma patients besides immune checkpoint inhibition. Since preclinical data suggests a synergistic effect of a MEK inhibitor (MEKi) and the oncolytic virus talimogene laherparepvec (T-VEC), we have treated three melanoma patients with this combination. All of the three patients had been suffering from recurring cutaneous and subcutaneous in-transit metastases. Upon treatment one patient (case 1) presented full regression of locoregional metastases and remained progression-free until date, for almost three years. The second patient (case 2) showed a partial regression of painful gluteal satellite metastases but died from brain metastases. The third patient (case 3) showed a durable response of locoregional metastases for seven months. The combination treatment was well tolerated with common adverse events known for each single agent. This report is the first case series presenting a clinical benefit of the combined T-VEC and MEKi treatment. We suggest the combination of T-VEC and MEKi as an off-label treatment option for patients with NRAS mutations, especially with recurrent in-transit or satellite metastases.
    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

       PDF(Pubmed)

  • 文章类型: Case Reports
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号