Oncogene Proteins, Fusion

癌基因蛋白质类,Fusion
  • 文章类型: Case Reports
    背景:原发性肺粘液样肉瘤(PPMS)是一种罕见的,低度恶性肿瘤,约占所有肺肿瘤的0.2%。尽管它很罕见,PPMS具有独特的组织学特征和分子改变,特别是EWSR1-CREB1基因融合的存在。然而,其精确的组织起源仍然难以捉摸,给临床诊断带来挑战。
    一名20岁男性患者在6个月前接受了常规体检,显示肺部肿块.手术切除后,微观评估揭示了主要是短纺锤形的肿瘤细胞组织在一个束状,梁状,或网状模式。基质基质显示出丰富的粘蛋白,伴有淋巴细胞和浆细胞浸润,拉塞尔的尸体在重点区域很明显。免疫表型分析显示肿瘤细胞中波形蛋白和上皮膜抗原阳性表达,而平滑肌肌动蛋白和S-100等,是阴性的。Ki-67增殖指数约为5%。随后的第二代测序鉴定了特征性的EWSR1-CREB1基因融合体。明确的病理诊断建立了PPMS。患者未接受辅助化疗或放疗,在30个月的随访期内仍无复发。
    结论:我们报告了一例位于左肺叶叶间裂内的罕见PPMS,以肿瘤间质内的罗素身体形成为特征,PPMS中的一个新发现。此外,这个病例的组织形态特征突出了它所带来的诊断挑战,因为它可能模仿炎性肌纤维母细胞瘤,骨外粘液样软骨肉瘤,或血管外皮细胞瘤样纤维组织细胞瘤。因此,准确的诊断需要一种涉及形态学的综合方法,免疫组织化学,和分子分析。
    BACKGROUND: Primary pulmonary myxoid sarcoma (PPMS) is a rare, low-grade malignant tumor, constituting approximately 0.2% of all lung tumors. Despite its rarity, PPMS possesses distinctive histological features and molecular alterations, notably the presence of EWSR1-CREB1 gene fusion. However, its precise tissue origin remains elusive, posing challenges in clinical diagnosis.
    UNASSIGNED: A 20-year-old male patient underwent a routine physical examination 6 months prior, revealing a pulmonary mass. Following surgical excision, microscopic evaluation unveiled predominantly short spindle-shaped tumor cells organized in a fascicular, beam-like, or reticular pattern. The stromal matrix exhibited abundant mucin, accompanied by lymphocytic and plasma cell infiltration, with Russell bodies evident in focal areas. Immunophenotypic profiling revealed positive expression of vimentin and epithelial membrane antigen in tumor cells, whereas smooth muscle actin and S-100, among others, were negative. Ki-67 proliferation index was approximately 5%. Subsequent second-generation sequencing identified the characteristic EWSR1-CREB1 gene fusion. The definitive pathological diagnosis established PPMS. The patient underwent no adjuvant chemotherapy or radiotherapy and remained recurrence-free during a 30-month follow-up period.
    CONCLUSIONS: We report a rare case of PPMS located within the left lung lobe interlobar fissure, featuring Russell body formation within the tumor stroma, a novel finding in PPMS. Furthermore, the histomorphological characteristics of this case highlight the diagnostic challenge it poses, as it may mimic inflammatory myofibroblastic tumor, extraskeletal myxoid chondrosarcoma, or hemangiopericytoma-like fibrous histiocytoma. Therefore, accurate diagnosis necessitates an integrated approach involving morphological, immunohistochemical, and molecular analyses.
    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
    靶向治疗和免疫治疗在非小细胞肺癌(NSCLC)的治疗中都很重要。准确的诊断和精确的治疗是实现患者长期生存的关键。MET融合是一种罕见的致癌因子,其最佳检测和治疗尚未建立。这里,我们报道了1例32岁女性肺腺癌患者,通过基于DNA的下一代测序(NGS)检测到PD-L1阳性和驱动基因阴性.化疗联合PD-1检查点抑制剂给药后根治性切除原发灶,根据她的病理反应和快速复发,表明原发性免疫抵抗。通过基于RNA的NGS检测到一种罕见的CD47-MET,荧光原位杂交证实了这一点。多重免疫荧光显示PD-L1相关的异质性免疫抑制微环境,几乎没有CD4T细胞和CD8T细胞分布。Savolitinib治疗导致无进展生存期(PFS)>12个月,直到疾病进展后通过重新活检和基于DNA-RNA的联合NGS在METp.D1228H中检测到新的继发性耐药突变.在这种情况下,CD47-MET融合非小细胞肺癌主要对免疫疗法耐药,对savolitinib敏感,靶向治疗后出现继发性METp.D1228H突变。基于DNA-RNA的NGS可用于检测此类分子事件和跟踪耐药性中的次级突变。为此,基于DNA-RNA的NGS可能在指导该患者人群的精确诊断和个体化治疗方面具有更好的价值。
    Targeted therapy and immunotherapy are both important in the treatment of non-small-cell lung cancer (NSCLC). Accurate diagnose and precise treatment are key in achieving long survival of patients. MET fusion is a rare oncogenic factor, whose optimal detection and treatment are not well established. Here, we report on a 32-year-old female lung adenocarcinoma patient with positive PD-L1 and negative driver gene detected by DNA-based next-generation sequencing (NGS). A radical resection of the primary lesion after chemotherapy combined with PD-1 checkpoint inhibitor administration indicated primary immuno-resistance according to her pathological response and rapid relapse. A rare CD47-MET was detected by RNA-based NGS, which was confirmed by fluorescence in situ hybridization. Multiplex immunofluorescence revealed a PD-L1 related heterogeneous immunosuppressive microenvironment with little distribution of CD4+ T cells and CD8+ T cells. Savolitinib therapy resulted in a progression-free survival (PFS) of >12 months, until a new secondary resistance mutation in MET p.D1228H was detected by re-biopsy and joint DNA-RNA-based NGS after disease progression. In this case, CD47-MET fusion NSCLC was primarily resistant to immunotherapy, sensitive to savolitinib, and developed secondary MET p.D1228H mutation after targeted treatment. DNA-RNA-based NGS is useful in the detection of such molecular events and tracking of secondary mutations in drug resistance. To this end, DNA-RNA-based NGS may be of better value in guiding precise diagnosis and individualized treatment in this patient population.
    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
    YAP1-KMT2A融合肉瘤是一种罕见的软组织肉瘤新亚型,好发于青壮年,形态学与硬化性上皮样纤维肉瘤(SEF)有一定程度重叠,但不表达MUC4。本例患者为34岁女性,左腋下无痛性肿块半年。肿瘤周界相对清楚,由大小形态相对一致的上皮样细胞呈小簇状、短条索状、梁状、巢状或弥漫片状分布于致密胶原化间质中,局部可见血管外皮瘤样结构和地图样坏死。瘤细胞无显著异型性,核分裂象约15个/10 HPF。瘤细胞表达上皮细胞膜抗原、CD99、cyclin D1和BCOR,MUC4阴性。荧光原位杂交显示KMT2A基因易位,二代测序显示YAP1-KMT2A基因融合。常规病理工作中,若遇到形态类似SEF,但不表达MUC4的肿瘤,要考虑到YAP1-KMT2A融合肉瘤的可能性,并行分子检测予以明确。.
    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: Case Reports
    暂无摘要。
    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: Case Reports
    先进分子技术的广泛使用已导致用PLAG1基因融合物鉴定了几种肿瘤类型,其中一些也影响皮肤和软组织。在这里,我们介绍了一名38岁的女性,她的前臂有皮下肿瘤,这似乎不适合任何当前公认的实体。这是一个界限清楚的肿瘤,大小为6×4,5×4厘米。它有一个厚厚的囊,由平淡的梭形细胞组成,在粘液胶原背景下形成栅栏和Verocay体样结构。分散的钙化分散在整个病变中。无细胞学异型,有丝分裂活性,或出现坏死。靶向NGS显示SOX10::PLAG1融合,荧光原位杂交证实了PLAG1基因重排的存在。肿瘤细胞显示S100,SOX10和PLAG1的弥漫性免疫组织化学表达,以及斑片状结蛋白和CD34阳性。该肿瘤的甲基化谱与DKFZ肉瘤分类器所覆盖的任何其他实体不匹配,并且除了染色体12的获得之外,拷贝数谱是正常的。肿瘤被完全切除,自切除以来,患者已经4年没有疾病了。虽然需要更多的病例来确认这种肿瘤是一个独特的实体,我们提出了一个临时名称“SOX10::PLAG1重排的钙化梭形细胞肿瘤。\"
    The widespread use of advanced molecular techniques has led to the identification of several tumor types with PLAG1 gene fusions some of which also affect the skin and soft tissues. Herein, we present a 38-year-old female with a subcutaneous tumor affecting her forearm, which does not seem to fit into any currently recognized entity. It was a well-circumscribed tumor measuring 6 × 4,5 × 4 cm. It had a thick capsule composed of bland spindle cells forming palisades and Verocay body-like structures within a myxocollagenous background. Scattered calcifications were dispersed throughout the lesion. No cytological atypia, mitotic activity, or necrosis were present. Targeted NGS revealed a SOX10::PLAG1 fusion and fluorescent in situ hybridization confirmed the presence of PLAG1 gene rearrangement. The neoplastic cells showed a diffuse immunohistochemical expression of S100, SOX10, and PLAG1, as well as patchy desmin and CD34 positivity. The methylation profile of this tumor did not match any other entity covered by the DKFZ sarcoma classifier and apart from the gain of chromosome 12, the copy number profile was normal. The tumor was completely excised, and the patient has been free of disease for 4 years since the excision. While more cases are needed to confirm this tumor as a distinct entity, we propose a provisional name \"SOX10::PLAG1-rearranged calcifying spindle cell tumor.\"
    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
    Lung cancer is the most common malignant disease and the leading cause of cancer death in China. Non-small cell lung cancer (NSCLC) accounts for over 80% of all lung cancers, and the probability of NSCLC gene mutations is high, with a wide variety of types. With the development of next-generation sequencing (NGS) detection technology, more and more patients with rare fusion gene mutations are detected. Neuregulin 1 (NRG1) gene is a rare oncogenic driver that can lead to activation of human epidermal growth factor receptor 3 (Her3/ErbB3) mediated pathway, resulting in tumor formation. In this article, we reported a case of mixed NSCLC with CRISPLD2-NRG1 fusion detected by RNA-based NGS, who responsed to Afatinib well after 1 month of treatment, and magnetic resonance imaging (MRI) showed shrinkage of intracranial lesions. Meanwhile, we also compiled previously reported NSCLC patients with NRG1 rare gene fusion mutation, in order to provide effective references for clinical diagnosis and treatment.
.
    【中文题目:罕见CRISPLD2-NRG1融合突变晚期混合型非小细胞肺癌1例并文献复习】 【中文摘要:肺癌是中国发病率和死亡率最高的恶性肿瘤。非小细胞肺癌(non-small cell lung cancer, NSCLC)占全部肺癌的80%以上,NSCLC的基因突变概率高,并且种类繁多。随着基因检测技术的进步,越来越多的罕见融合基因变异被检测出来。神经调节蛋白1(neuregulin 1, NRG1)可促使人表皮生长因子受体3(human epidermal growth factor receptor 3, Her3/ErbB3)介导的通路激活,从而导致肿瘤形成。本文报道了1例罕见CRISPLD2-NRG1融合突变的晚期混合型NSCLC颅内转移的患者,接受阿法替尼治疗1个月后头部磁共振成像(magnetic resonance imaging, MRI)显示颅内病灶明显缩小,患者对阿法替尼治疗反应良好。同时,我们对以往报道的NRG1基因融合突变的NSCLC病例进行总结,以供临床借鉴。
】 【中文关键词:肺肿瘤;阿法替尼;CRISPLD2-NRG1融合突变】.
    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

    求助全文

  • 文章类型: Letter
    暂无摘要。
    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
    NUT Carcinoma(NC) is a rare malignant tumor of unknown origin, which is highly aggressive. It is characterized by chromosome rearrangement accompanied by NUTM1 gene. The pathological manifestations were sudden and focal squamous in poorly differentiated or undifferentiated carcinoma. NUTM1gene rearrangement can be used to diagnose NC. The prognosis of NUT cancer is poor. Clinically, there is no established treatment plan. treatment options mainly comprise surgery, radiotherapy and chemotherapy. A 74-year-old patient with NC of the nasal cavity and sinuses was reported. Her clinical presentation was right nasal congestion with facial swelling. Sinus CT and MRI showed soft tissue density in the right nasal cavity and maxillary sinus with bone destruction. After admission, the patient underwent nasal endoscopic biopsy, and the postoperative pathological FISH staining showed BRD4/NUT fusion t(15, 19). The tumor was significantly reduced after two courses of sequential chemoradiotherapy. Two months later, the patient underwent a partial maxillary resection due to the rapid regrowth of sinusoidal mass, invading the hard palate. The patient died 2 months after surgery due to multiple organ failure resulted from tumor metastasis, with a survival time of 11 months. The clinical characteristics, diagnosis and treatment of this case were reported and related literature was reviewed.
    摘要: NUT癌(NUT Carcinoma)是一种罕见的起源不明的具有高度侵袭性的恶性肿瘤,其以伴有NUTM1基因染色体重排为主要特点,病理表现为具有突然和局灶性鳞状分化低分化或未分化癌,FISH检测见NUTM1基因重排可明确诊断,NUT癌预后较差,临床上并未明确治疗方案,治疗方式多为手术、放疗及化疗。本文报道1例74岁鼻腔鼻窦NUT癌患者,临床表现为右侧鼻塞伴面部肿胀,鼻窦CT及MRI示右侧鼻腔及上颌窦软组织密度影伴骨质破坏,入院后行鼻窦肿物活检术,术后病理FISH染色结果示BRD4/NUT融合t(15,19),予序贯同步放化疗2个疗程后肿物显著减小,2个月后肿物再次迅速增长侵及硬腭影响进食,行上颌骨部分切除术,术后2个月患者因肿瘤转移累及全身脏器衰竭死亡,生存期11个月,现对本病例的临床特征和诊疗经过进行报告及相关文献复习。.
    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
    在世界卫生组织最新的第五版中枢神经系统肿瘤分类中,星形母细胞瘤已被定义为涉及MN1基因的分子重排,与共同的合作伙伴是BEND2或CXXC5。因此,这个肿瘤实体现在被称为“星形母细胞瘤”,MN1-改变。\"然而,EWSR1胶质瘤::BEND2融合,没有MN1融合改变,最近已被证明表现出星形母细胞瘤样组织形态学特征,并驻留在一个独特的表观遗传亚组,基于DNA甲基化研究,类似于具有MN1改变的高级别神经上皮肿瘤,其中包括星形母细胞瘤,MN1改变肿瘤。包含EWSR1的星形母细胞瘤的这种新的表观遗传独特亚型::BEND2融合缺乏所需的MN1改变,并且,因此,不满足目前这些病变的分子分类。这里,我们描述了1例胶质瘤,其组织学特征和DNA甲基化分析与新型YAP1::BEND2融合的星形母细胞瘤一致.这种情况和其他情况进一步扩大了在MN1改变的限制之外的星形母细胞瘤样肿瘤中可观察到的分子发现。此类EWSR1::BEND2和YAP1::BEND2融合的星形母细胞瘤病例挑战了仅基于MN1改变的星形母细胞瘤的当前分子分类。
    In the most recent fifth edition of the World Health Organization Classification of Tumors of the Central Nervous System, astroblastoma has been defined by molecular rearrangements involving the MN1 gene, with common partners being BEND2 or CXXC5 . Accordingly, this tumor entity is now known as \"astroblastoma, MN1 -altered.\" However, gliomas with EWSR1::BEND2 fusions, devoid of MN1 fusion alterations, have recently been shown to exhibit astroblastoma-like histomorphologic features and reside in a distinct epigenetic subgroup based on DNA methylation studies similar to high-grade neuroepithelial tumor with MN1 alteration, which includes astroblastoma, MN1 altered tumors. This new epigenetically distinct subtype of astroblastoma containing EWSR1::BEND2 fusions lacks the required MN1 alteration and, thus, does not satisfy the current molecular classification of these lesions. Here, we describe a case of glioma with histologic features and DNA methylation profiling consistent with astroblastoma with a novel YAP1: : BEND2 fusion. This case and others further expand the molecular findings observable in astroblastoma-like tumors outside the constraints of MN1 alteration. Such cases of astroblastoma with EWSR1::BEND2 and YAP1::BEND2 fusions challenge the current molecular classification of astroblastoma based solely on an MN1 alteration.
    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