T-VEC

T - VEC
  • 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: 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: Case Reports
    NUT癌(NC)是一种罕见且极具侵袭性的癌症,通常在青少年和年轻人中表现为胸内或颈部表现。没有既定的标准治疗方案,中位总生存期仅为6.5个月,有一个巨大的需要创新的治疗方案。由于NC是由单个异常融合癌蛋白遗传驱动的,它通常以肿瘤突变负担低为特征,从而使其免疫寒冷,对常规免疫疗法不敏感。最近,我们已经证明,溶瘤病毒(OVs)能够特异性感染和裂解NC细胞,从而将免疫冷的肿瘤微环境变成热的。这里,我们报告了一种强化的多模式治疗方法,首次采用OV(talimogenelaherparepvec(T-VEC);IMLYGIC®)与免疫检查点抑制剂pembrolizumab作为基础NC治疗的附加药物(细胞抑制化疗,放射治疗,表观遗传治疗)在患有大的胸部NC肿瘤的患者中表现出异常,独特的BRD3:NUTM1融合。这个案例首次证明了这种创新的附加免疫病毒治疗方案的可行性,T-VEC的重复和持久复制有助于实现肿瘤稳定和改善患者的生活质量。Further,一个以前未知的BRD3:NUTM1融合基因被发现缺乏BRD3的外结构域。
    NUT carcinoma (NC) is a rare and extremely aggressive form of cancer, usually presenting with intrathoracic or neck manifestations in adolescents and young adults. With no established standard therapy regimen and a median overall survival of only 6.5 months, there is a huge need for innovative treatment options. As NC is genetically driven by a single aberrant fusion oncoprotein, it is generally characterized by a low tumor mutational burden, thus making it immunologically cold and insusceptible to conventional immunotherapy. Recently, we have demonstrated that oncolytic viruses (OVs) are able to specifically infect and lyse NC cells, thereby turning an immunologically cold tumor microenvironment into a hot one. Here, we report an intensive multimodal treatment approach employing for the first time an OV (talimogene laherparepvec (T-VEC); IMLYGIC®) together with the immune checkpoint inhibitor pembrolizumab as an add-on to a basic NC therapy (cytostatic chemotherapy, radiation therapy, epigenetic therapy) in a patient suffering from a large thoracic NC tumor which exhibits an aberrant, unique BRD3:NUTM1 fusion. This case demonstrates for the first time the feasibility of this innovative add-on immunovirotherapy regimen with a profound, repetitive and durable replication of T-VEC that is instrumental in achieving tumor stabilization and improvement in the patient´s quality of life. Further, a previously unknown BRD3:NUTM1 fusion gene was discovered that lacks the extraterminal domain of BRD3.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号