Oncogene Proteins, Fusion

癌基因蛋白质类,Fusion
  • 文章类型: Journal Article
    背景:神经营养酪氨酸受体激酶(NTRK)基因融合是致癌驱动因素。使用芬兰的Auria生物库,我们的目的是识别和表征这些基因融合的患者,并描述它们的临床和肿瘤特征,接受治疗,和结果。
    方法:我们评估了任何实体瘤类型的儿科和成人结直肠癌(CRC),非小细胞肺癌(NSCLC),肉瘤,或者唾液腺癌.我们通过对AuriaBiobank的肿瘤样品进行pan-TRK免疫组织化学(IHC)染色,确定了原肌球蛋白受体激酶(TRK)蛋白的表达,由认证的病理学家评分。通过下一代测序(NGS)确认NTRK基因融合。所有2,059名患者在癌症诊断前1年开始随访。
    结果:儿科NTRK基因融合瘤的发生率为3.1%(4/127),CRC的0.7%(8/1,151),非小细胞肺癌0.3%(1/288),0.9%(1/114)为唾液腺癌,0%(0/379)为肉瘤。在儿科中,纤维肉瘤各1例(TPM3::NTRK1),尤因肉瘤(LPPR1::NTRK2),原始神经外胚层肿瘤(DAB2IP::NTRK2),甲状腺乳头状癌(RAD51B::NTRK3)。在CRC患者中,六个带有NTRK1融合的肿瘤(三个与TPM3融合),一个人拥有NTRK3::GABRG1融合,另一种是NTRK2::FXN/LPPR1融合体。与野生型肿瘤相比,NTRK基因融合肿瘤的CRC患者的微卫星不稳定性更高(50.0%vs.4.4%)。其他检测到的融合是SGCZ::NTRK3(NSCLC)和ETV6::NTRK3(唾液腺癌)。四名患者(三名CRC,1例NSCLC)接受化疗;1例(CRC)接受放疗。
    结论:NTRK基因融合在成人CRC中很少见,NSCLC,唾液肿瘤,肉瘤,和小儿实体瘤。
    BACKGROUND: Neurotrophic tyrosine receptor kinase (NTRK) gene fusions are oncogenic drivers. Using the Auria Biobank in Finland, we aimed to identify and characterize patients with these gene fusions, and describe their clinical and tumor characteristics, treatments received, and outcomes.
    METHODS: We evaluated pediatrics with any solid tumor type and adults with colorectal cancer (CRC), non-small cell lung cancer (NSCLC), sarcoma, or salivary gland cancer. We determined tropomyosin receptor kinase (TRK) protein expression by pan-TRK immunohistochemistry (IHC) staining of tumor samples from the Auria Biobank, scored by a certified pathologist. NTRK gene fusion was confirmed by next generation sequencing (NGS). All 2,059 patients were followed-up starting 1 year before their cancer diagnosis.
    RESULTS: Frequency of NTRK gene fusion tumors was 3.1% (4/127) in pediatrics, 0.7% (8/1,151) for CRC, 0.3% (1/288) for NSCLC, 0.9% (1/114) for salivary gland cancer, and 0% (0/379) for sarcoma. Among pediatrics there was one case each of fibrosarcoma (TPM3::NTRK1), Ewing\'s sarcoma (LPPR1::NTRK2), primitive neuroectodermal tumor (DAB2IP::NTRK2), and papillary thyroid carcinoma (RAD51B::NTRK3). Among CRC patients, six harbored tumors with NTRK1 fusions (three fused with TPM3), one harbored a NTRK3::GABRG1 fusion, and the other a NTRK2::FXN/LPPR1 fusion. Microsatellite instability was higher in CRC patients with NTRK gene fusion tumors versus wild-type tumors (50.0% vs. 4.4%). Other detected fusions were SGCZ::NTRK3 (NSCLC) and ETV6::NTRK3 (salivary gland cancer). Four patients (three CRC, one NSCLC) received chemotherapy; one patient (with CRC) received radiotherapy.
    CONCLUSIONS: NTRK gene fusions are rare in adult CRC, NSCLC, salivary tumors, sarcoma, and pediatric solid tumors.
    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
    神经营养性酪氨酸受体激酶(NTRK)基因融合是在0.3%的实体瘤中普遍存在的罕见致癌驱动因素。它们在唾液腺癌中最常见(2.6%),甲状腺癌(1.6%),软组织肉瘤(1.5%)。目前,有两种美国食品和药物管理局批准的NTRK基因融合的靶向疗法:larotrectinib,2018年批准,和entrectinib,2019年批准。迄今为止,在学术癌症中心中,酪氨酸受体激酶抑制剂(TRKi)在NTRK阳性实体瘤中的实际使用情况仍在很大程度上未知.
    为了描述人口统计,临床和基因组特征,以及在美国学术癌症中心治疗的NTRK阳性实体瘤患者的测试和治疗模式。
    这是一项在美国学术癌症中心进行的回顾性图表综述研究。本研究包括所有诊断为NTRK融合阳性(NTRK1,NTRK2,NTRK3)实体瘤(任何阶段)并且在2012年1月1日至2023年7月1日期间在参与部位接受癌症治疗的患者。患者人口统计学,临床特征,基因组特征,NTRK测试数据,从电子病历中收集治疗模式,并酌情使用描述性统计进行分析.
    总共,6个中心提供了55例NTRK阳性肿瘤患者的数据。平均年龄为49.3(SD=20.5)岁,51%的患者为女性,大多数是白人(78%)。从癌症诊断到NTRK测试的中位持续时间为85天(IQR=44-978)。在NTRK测试时,64%的患者患有IV期疾病,与癌症诊断时的33%相比。总体队列中流行的癌症类型包括头颈部(15%),甲状腺(15%),大脑(13%),肺(13%),结直肠(11%)。NTRK1融合最常见(45%),其次是NTRK3(40%)和NTRK2(15%)。在所有的治疗中,51%(n=28)的患者接收了TRKi。在TRKi治疗的患者中,71%的人在TRKi开始时患有IV期疾病。从NTRK试验阳性到开始TRKi的中位时间为48天(IQR=9-207)。TRKis通常作为一线(30%)或二线(48%)疗法给予。使用TRKi的中位治疗持续时间为610天(IQR=182-764),所有其他一线治疗的中位治疗持续时间为207.5天(IQR=42-539)。
    这项研究报告了当代现实世界的NTRK测试模式和TRKis在实体瘤中的应用,包括NTRK测试与开始TRKi治疗之间的时间以及TRKi治疗的持续时间。
    UNASSIGNED: Neurotrophic tyrosine receptor kinase (NTRK) gene fusions are rare oncogenic drivers prevalent in 0.3% of solid tumors. They are most common in salivary gland cancer (2.6%), thyroid cancer (1.6%), and soft-tissue sarcoma (1.5%). Currently, there are 2 US Food and Drug Administration-approved targeted therapies for NTRK gene fusions: larotrectinib, approved in 2018, and entrectinib, approved in 2019. To date, the real-world uptake of tyrosine receptor kinase inhibitor (TRKi) use for NTRK-positive solid tumors in academic cancer centers remains largely unknown.
    UNASSIGNED: To describe the demographics, clinical and genomic characteristics, and testing and treatment patterns of patients with NTRK-positive solid tumors treated at US academic cancer centers.
    UNASSIGNED: This was a retrospective chart review study conducted in academic cancer centers in the United States. All patients diagnosed with an NTRK fusion-positive (NTRK1, NTRK2, NTRK3) solid tumor (any stage) and who received cancer treatment at participating sites between January 1, 2012, and July 1, 2023, were included in this study. Patient demographics, clinical characteristics, genomic characteristics, NTRK testing data, and treatment patterns were collected from electronic medical records and analyzed using descriptive statistics as appropriate.
    UNASSIGNED: In total, 6 centers contributed data for 55 patients with NTRK-positive tumors. The mean age was 49.3 (SD = 20.5) years, 51% patients were female, and the majority were White (78%). The median duration of time from cancer diagnosis to NTRK testing was 85 days (IQR = 44-978). At the time of NTRK testing, 64% of patients had stage IV disease, compared with 33% at cancer diagnosis. Prevalent cancer types in the overall cohort included head and neck (15%), thyroid (15%), brain (13%), lung (13%), and colorectal (11%). NTRK1 fusions were most common (45%), followed by NTRK3 (40%) and NTRK2 (15%). Across all lines of therapy, 51% of patients (n = 28) received a TRKi. Among TRKi-treated patients, 71% had stage IV disease at TRKi initiation. The median time from positive NTRK test to initiation of TRKi was 48 days (IQR = 9-207). TRKis were commonly given as first-line (30%) or second-line (48%) therapies. Median duration of therapy was 610 (IQR = 182-764) days for TRKi use and 207.5 (IQR = 42-539) days for all other first-line therapies.
    UNASSIGNED: This study reports on contemporary real-world NTRK testing patterns and use of TRKis in solid tumors, including time between NTRK testing and initiation of TRKi therapy and duration of TRKi 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: 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: Journal Article
    背景:虽然融合驱动的软组织肿瘤正在迅速扩大,它们在皮肤和浅层间充质和附件肿瘤中的重要性仍然知之甚少。这种挑战在具有难以基于形态学进行分类的不明确的组织病理学的情况下尤其明显。
    目的:我们的目标是研究下一代测序在诊断复杂皮肤肿瘤中的益处。
    方法:在部门档案中搜索融合驱动的皮肤肿瘤。检索载玻片并获得包括随访的临床信息。
    结果:15例发生在8名女性和7名男性患者中,诊断时的中位年龄为26岁(范围:1-83)。肿瘤累及四肢(9),头皮(5),头部和颈部(1)。主要特征包括肌上皮(5),巢状,细胞质清晰(2),非典型附件/鳞状(2),小圆形蓝色细胞(2),细胞纺锤状(3),和纤维组织细胞形态(1)。最常见的融合涉及EWSR1(6)与ERG(1)融合,FLI1(1),CREB1(2),CREM(1),PBX3(1),其次是PLAG1(4)和LIFR(2),TRPS1(1)和CHCHD7。遇到的其他融合是YAP1::NUTM1,EML4::ALK,SS18::SSX1(2),和一个新的融合:ACTB::ZMIZ2。整合组织学特征和分子表现导致原发性皮肤尤文肉瘤的最终诊断(2),软组织肌上皮瘤(4),皮肤合胞肌上皮瘤(1),皮肤附件癌(1),胃癌(1),炎性肌纤维母细胞瘤(1),滑膜肉瘤(2),透明细胞肉瘤(2),血管瘤样纤维组织细胞瘤(1)。
    结论:我们的结果表明,融合测试可能是一种有用的诊断工具,特别是在表面部位有异常或罕见形态的病例。此外,在某些情况下,它可以识别潜在的治疗靶标。
    BACKGROUND: While the list of fusion-driven soft tissue neoplasms is expanding rapidly, their importance among cutaneous and superficial mesenchymal and adnexal neoplasms remains poorly understood. This challenge is especially evident in cases with ambiguous histopathology that are difficult to classify based on morphology.
    OBJECTIVE: Our goal was to investigate the benefits of next-generation sequencing in diagnosing complex cutaneous neoplasms.
    METHODS: Departmental archives were searched for fusion-driven cutaneous neoplasms. Slides were retrieved and clinical information including follow-up was obtained.
    RESULTS: Fifteen cases occurred in eight female and seven male patients, with a median age of 26 years (range: 1-83) at diagnosis. Tumors involved the extremities (9), scalp (5), and head and neck (1). Predominant features included myoepithelial (5), nested spindled with clear cytoplasm (2), atypical adnexal/squamoid (2), small round blue cell (2), cellular spindled (3), and fibrohistiocytic morphology (1). Most frequently encountered fusions involved EWSR1 (6) fused to ERG (1), FLI1 (1), CREB1 (2), CREM (1), PBX3 (1), followed by PLAG1 (4) with LIFR (2), TRPS1 (1) and CHCHD7. Additional fusions encountered were YAP1::NUTM1, EML4::ALK, SS18::SSX1 (2), and a novel fusion: ACTB::ZMIZ2. Integration of histologic features and molecular findings led to final diagnoses of primary cutaneous Ewing sarcoma (2), soft tissue myoepithelioma (4), cutaneous syncytial myoepithelioma (1), cutaneous adnexal carcinoma (1), porocarcinoma (1), inflammatory myofibroblastic tumor (1), synovial sarcoma (2), clear cell sarcoma (2), and angiomatoid fibrous histiocytoma (1).
    CONCLUSIONS: Our results show that fusion testing can be a helpful diagnostic tool, especially in cases with unusual or uncommon morphology in superficial sites. Furthermore, it can allow for the identification of potential therapeutic targets in some instances.
    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
    背景:本研究的目的是探讨临床因素与FOXO1融合状态在非转移性横纹肌肉瘤(RMS)患者中的作用,以建立无事件生存的预测模型,并为未来试验的风险分层提供依据。
    方法:作者使用了来自欧洲儿童软组织肉瘤研究组(EpSSG)RMS2005研究(EpSSGRMS2005;EudraCT编号2005-000217-35)的患者的数据。多变量模型考虑了以下基线变量:诊断时的年龄,性别,组织学,原发肿瘤部位,组间横纹肌肉瘤研究组,肿瘤大小,节点状态,和FOXO1融合状态。候选预测因子的主要效应和显著的二阶相互作用被纳入多重Cox比例风险回归模型。生成用于预测5年无事件生存(EFS)概率的列线图。
    结果:5年EFS和总生存率为70.9%(95%置信区间,68.6%-73.1%)和81.0%(95%置信区间,78.9%-82.8%),分别。多变量模型保留了五个预后因素,包括诊断时的年龄与肿瘤大小的相互作用,肿瘤原发部位,组间横纹肌肉瘤研究临床组,和FOXO1融合状态。根据列线图中每个病人的总分,患者被分为四组.5年EFS率为94.1%,78.4%,65.2%,和52.1%的低风险,中等风险,高风险,高危人群,分别,相应的5年总生存率为97.2%,91.5%,74.3%,和60.8%,分别。
    结论:此处提供的结果提供了修改EpSSG分层的基本原理,以融合状态取代组织学为代表的最显著变化。EpSSG发起的新国际试验采用了这种分类。
    BACKGROUND: The objective of this study was to investigate the role of clinical factors together with FOXO1 fusion status in patients with nonmetastatic rhabdomyosarcoma (RMS) to develop a predictive model for event-free survival and provide a rationale for risk stratification in future trials.
    METHODS: The authors used data from patients enrolled in the European Pediatric Soft Tissue Sarcoma Study Group (EpSSG) RMS 2005 study (EpSSG RMS 2005; EudraCT number 2005-000217-35). The following baseline variables were considered for the multivariable model: age at diagnosis, sex, histology, primary tumor site, Intergroup Rhabdomyosarcoma Studies group, tumor size, nodal status, and FOXO1 fusion status. Main effects and significant second-order interactions of candidate predictors were included in a multiple Cox proportional hazards regression model. A nomogram was generated for predicting 5-year event-free survival (EFS) probabilities.
    RESULTS: The EFS and overall survival rates at 5 years were 70.9% (95% confidence interval, 68.6%-73.1%) and 81.0% (95% confidence interval, 78.9%-82.8%), respectively. The multivariable model retained five prognostic factors, including age at diagnosis interacting with tumor size, tumor primary site, Intergroup Rhabdomyosarcoma Studies clinical group, and FOXO1 fusion status. Based on each patient\'s total score in the nomogram, patients were stratified into four groups. The 5-year EFS rates were 94.1%, 78.4%, 65.2%, and 52.1% in the low-risk, intermediate-risk, high-risk, and very-high-risk groups, respectively, and the corresponding 5-year overall survival rates were 97.2%, 91.5%, 74.3%, and 60.8%, respectively.
    CONCLUSIONS: The results presented here provide the rationale to modify the EpSSG stratification, with the most significant change represented by the replacement of histology with fusion status. This classification was adopted in the new international trial launched by the EpSSG.
    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
    目的:TFE3重排的肾细胞癌(RCC)在TFE3和许多伴侣基因中的1之间存在基因融合。MED15::TFE3融合RCC是罕见的,通常是囊性的,容易误诊。
    方法:本研究旨在通过荧光原位杂交和靶向RNA测序来表征2例具有广泛囊性改变的MED15::TFE3融合RCC。
    结果:两名患者均为29岁和35岁的年轻成年女性。放射学上,两者均表现为囊性BosniakII类肾脏病变。囊肿的最大尺寸为9.3厘米和4.8厘米。两名患者都接受了囊肿摘除术,随访26个月和6个月均无肿瘤复发或转移,分别。微观上,两种肿瘤都是完全囊性的,厚厚的,纤维囊壁内衬小细胞簇,细胞质清晰至嗜酸性细胞,均匀,核仁不明显的圆形核。囊壁内也有类似透明细胞的小聚集。在1例中发现了基底膜样物质沉积的病灶;在两种情况下都观察到钙化。两例均显示PAX8和TFE3的核阳性和Melan-A的细胞质染色;HMB45,CAIX,CK7为阴性。荧光原位杂交显示两个肿瘤对TFE3重排均为阳性。RNA测序在两种情况下都鉴定了MED15::TFE3基因融合体。
    结论:MED15::TFE3融合肾癌的主要鉴别诊断包括低恶性潜能的多房性囊性肾肿瘤和不典型的肾囊肿。TFE3融合的分子确认对于建立正确的诊断至关重要。
    OBJECTIVE: TFE3-rearranged renal cell carcinomas (RCCs) harbor gene fusions between TFE3 and 1 of many partner genes. MED15::TFE3 fusion RCC is rare, often cystic, and easily misdiagnosed.
    METHODS: This study aimed to characterize 2 cases of MED15::TFE3 fusion RCC with extensive cystic change using fluorescence in situ hybridization and targeted RNA sequencing.
    RESULTS: Both patients were young adult women aged 29 and 35 years. Radiologically, both presented with a cystic Bosniak category II renal lesion. The cysts measured 9.3 cm and 4.8 cm in greatest dimension. Both patients underwent cyst enucleation, and neither had tumor recurrence or metastasis at 26 and 6 months of follow-up, respectively. Microscopically, both tumors were entirely cystic, with thick, fibrous cystic walls lined by small clusters of cells with clear to eosinophilic cytoplasm and uniform, round nuclei with inconspicuous nucleoli. There were also small aggregations of similar clear cells within the cystic walls. Foci of basement membrane-like material depositions were noted in 1 case; calcifications were observed in both cases. Both cases demonstrated nuclear positivity for PAX8 and TFE3 and cytoplasmic staining for Melan-A; HMB45, CAIX, and CK7 were negative. Fluorescence in situ hybridization revealed that both tumors were positive for TFE3 rearrangements. RNA sequencing identified MED15::TFE3 gene fusions in both cases.
    CONCLUSIONS: The main differential diagnosis of MED15::TFE3 fusion RCC includes multilocular cystic renal neoplasm of low malignant potential and atypical renal cysts. Molecular confirmation of TFE3 fusion is essential for establishing the correct diagnosis.
    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
    神经调节素1(NRG1)融合是在非小细胞肺癌(NSCLC)中检测到的致癌驱动因素,胰腺导管腺癌(PDAC)和其他实体瘤。NRG1融合是罕见的,发生在不到1%的实体瘤中。NRG1融合阳性(NRG1)癌症患者的治疗选择有限。Zenocutuzumab是一种新颖的,双特异性IgG1抗体,靶向HER2和HER3蛋白并通过“Dock&Block®”作用机制抑制NRG1结合。这里,我们描述了eNRGy试验第二阶段组成部分的基本原理和设计,整体的一部分,开放标签阶段I/II,探索安全性的多中心试验,耐受性,药代动力学,药效学,zenocutuzumab在NRG1+NSCLC患者中的免疫原性和抗肿瘤活性,PDAC或其他实体瘤。
    NRG1基因融合是导致癌细胞生长的罕见突变。这些融合在许多不同类型的癌症中发现。具有NRG1基因融合的肿瘤对标准治疗方案的反应不佳。Zenocutuzumab,或者芝诺,是一种正在测试的治疗方法,看看它是否可以阻止由于NRG1基因融合而生长的癌症。这里,我们描述了一项正在进行的临床试验(eNRGy)的推理和设计,该试验旨在研究Zeno在NRG1基因融合癌症患者中的疗效(效果)和安全性.eNRGy试验正在招募具有NRG1基因融合的癌症患者,包括非小细胞肺癌,胰腺癌和其他。参加这项试验的患者将每两周接受一次Zeno,直到他们的癌症生长。该试验的主要目标(主要终点)是确定肿瘤大小减少30%或更多的患者的百分比。eNRGy试验目前正在招募患者。有关更多信息,参考ClinicalTrials.gov(标识符:NCT02912949),访问https://nrg1.com/,或致电1-833-NRG-1234。
    Neuregulin 1 (NRG1) fusions are oncogenic drivers that have been detected in non-small-cell lung cancer (NSCLC), pancreatic ductal adenocarcinoma (PDAC) and other solid tumors. NRG1 fusions are rare, occurring in less than 1% of solid tumors. Patients with NRG1 fusion positive (NRG1+) cancer have limited therapeutic options. Zenocutuzumab is a novel, bispecific IgG1 antibody that targets both HER2 and HER3 proteins and inhibits NRG1 binding through a \'Dock & Block®\' mechanism of action. Here, we describe the rationale and design of the phase II component of the eNRGy trial, part of the overall, open-label phase I/II, multicenter trial exploring the safety, tolerability, pharmacokinetics, pharmacodynamics, immunogenicity and antitumor activity of zenocutuzumab in patients with NRG1+ NSCLC, PDAC or other solid tumors.
    eNRGy: a clinical trial of zenocutuzumab for cancer caused by NRG1 gene fusionsNRG1 gene fusions are rare mutations that cause cancer cells to grow. These fusions are found in many different types of cancer. Tumors with NRG1 gene fusions do not respond well to standard treatment options. Zenocutuzumab, or Zeno, is a treatment that is being tested to see if it can stop cancer that is growing because of NRG1 gene fusions. Here, we describe the reasoning for and design of an ongoing clinical trial (eNRGy) designed to study the efficacy (how well it works) and safety of Zeno in patients with cancer that has NRG1 gene fusions. The eNRGy trial is recruiting patients with cancer that has NRG1 gene fusions, including non-small-cell lung cancer, pancreatic cancer and others. Patients who join this trial will receive Zeno once every 2 weeks until their cancer grows. The main goal (primary end point) of this trial is to determine the percentage of patients whose tumors decrease in size by 30% or more. The eNRGy trial is currently enrolling patients. For more information, refer to ClinicalTrials.gov (Identifier: NCT02912949), visit https://nrg1.com/, or call 1-833-NRG-1234.
    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
    频率,在ALK阳性间变性大细胞淋巴瘤(ALCL)中除NPM1以外的ALK伴侣基因的分布和预后意义尚不清楚.在NHL-BFM研究组中诊断出的316个ALCL中有49个显示没有暗示变体ALK伴侣的核ALK表达;通过基因组捕获高通量测序或特异性RT-PCR分析了41个。13例检出NPM1::ALK。在28例非NPM1::ALK融合伴侣的患者中,ATIC(n=8;29%)和TPM3(n=9;32%)最常见。8例ATIC患者中有5例::ALK阳性ALCL复发,九个都没有TPM3::ALK。变异的ALK伴侣很少见,可能与不同的预后相关。
    Frequency, distribution and prognostic meaning of ALK-partner genes other than NPM1 in ALK-positive anaplastic large-cell lymphoma (ALCL) are unknown. Forty-nine of 316 ALCL diagnosed in the NHL-BFM study group showed no nuclear ALK expression suggestive of a variant ALK-partner; 41 were analysed by genomic capture high-throughput sequencing or specific RT-PCRs. NPM1::ALK was detected in 13 cases. Among the 28 patients with a non-NPM1::ALK-fusion partner, ATIC (n = 8; 29%) and TPM3 (n = 9; 32%) were the most common. Five of eight patients with ATIC::ALK-positive ALCL relapsed, none of nine with TPM3::ALK. Variant ALK-partners are rare and potentially associated with different prognoses.
    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
    描述了7例原发性肺肿瘤,其组织学特征为透明细胞形态和独特的FGFR3::TACC3基因重排。肿瘤出现在4名女性和3名男性中,年龄47至81岁(平均=68)。它们发生在外围位置,主要是胸膜下,大小范围从1.4到6.5厘米(平均=4.1厘米)。所有肿瘤均表现出稳固的生长模式,具有丰富的中央坏死区域和明显的核多态性。肿瘤显示透明细胞组织学,具有大的粘性肿瘤细胞,显示出非典型的细胞核和丰富的透明细胞质。免疫组织化学染色鉴定5例鳞状表型和2例腺癌表型。一例是鳞状细胞癌,伴有局灶性腺体成分,其中一个鳞状细胞癌显示局灶性肉瘤样改变。下一代测序在所有7例中鉴定了FGFR3::TACC3基因重排。一个病例显示了同时激活的FGFR3突变,第二个病例显示了同时FGFR3扩增。2例并发KRASG12D突变。一例同时存在KRAS和EGFR突变,1例并发TP53突变。携带FGFR3::TACC3基因重排的非小细胞肺癌极为罕见,并且这种重排可能潜在地富集在显示清晰细胞组织学的肿瘤中。在具有透明细胞特征的肺癌患者中鉴定FGFR3::TACC3可能很重要,因为它们可能是酪氨酸激酶抑制剂治疗的候选药物。
    Seven cases of primary lung tumors characterized histologically by clear cell morphology and a distinctive FGFR3::TACC3 gene rearrangement are described. The tumors arose in 4 women and 3 men, aged 47 to 81 years (mean=68). They occurred in peripheral locations, predominantly subpleural, and ranged in size from 1.4 to 6.5 cm (mean=4.1 cm). All tumors showed a solid growth pattern with abundant central areas of necrosis and marked nuclear pleomorphism. The tumors demonstrated clear cell histology, with large cohesive tumor cells displaying atypical nuclei and abundant clear cytoplasm. Immunohistochemical stains identified a squamous phenotype in 5 cases and an adenocarcinoma phenotype in 2 cases. One case was a squamous cell carcinoma with focal glandular component, and one of the squamous cell carcinomas showed focal sarcomatoid changes. Next generation sequencing identified FGFR3::TACC3 gene rearrangements in all 7 cases. One case demonstrated a concurrent activating FGFR3 mutation and a second case demonstrated concurrent FGFR3 amplification. Two cases harbored a concurrent KRAS G12D mutation. One case harbored both KRAS and EGFR mutations, and 1 case had a concurrent TP53 mutation. Non-small cell lung carcinoma harboring FGFR3::TACC3 gene rearrangements is extremely rare, and this rearrangement may potentially be enriched in tumors that demonstrate clear cell histology. Identification of FGFR3::TACC3 in patients with lung carcinomas with clear cell features may be of importance as they could potentially be candidates for therapy with tyrosine kinase inhibitors.
    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