BRAF fusion

  • 文章类型: Journal Article
    自从发现BRAF突变和融合在儿童低级别神经胶质瘤发展中的关联以及随后认识到大多数儿童低级别神经胶质和神经胶质细胞肿瘤通过RAS/RAF/MAP激酶途径具有异常信号传导以来,这些肿瘤的概念化方式发生了巨大的变化。这些肿瘤中存在的许多融合和突变与分子靶标有关,有正在开发或已经在临床使用的药物。各种代理商,包括MEK抑制剂,BRAF抑制剂,MTOR抑制剂和,在NTRK抑制剂的小亚群患者中,已成功用于治疗复发性疾病的儿童,在常规方法如手术或化疗失败后。与新诊断的神经胶质瘤和神经胶质肿瘤儿童的分子靶向治疗相比,化疗的相对益处正在研究中。在新诊断的BRAF-V600E突变的低度神经胶质瘤和神经胶质肿瘤儿童中,MEK抑制剂和BRAF抑制剂的组合已显示优于常规化疗(卡铂和长春新碱)。然而,这种分子靶向治疗的长期效果尚不清楚.分子靶向治疗在早期治疗中的潜在用途使得在开始治疗之前必须知道大多数低级别神经胶质和神经胶质细胞肿瘤的分子组成。该规则的主要例外是患有1型神经纤维瘤病的儿童,根据定义,有NF1损失;然而,即使在这个人群中,出现在儿童晚期和青春期或对常规治疗无反应的胶质瘤可能是活检的候选人,尤其是在进入分子靶向治疗试验之前。
    Since the discovery of the association between BRAF mutations and fusions in the development of childhood low-grade gliomas and the subsequent recognition that most childhood low-grade glial and glioneuronal tumors have aberrant signaling through the RAS/RAF/MAP kinase pathway, there has been a dramatic change in how these tumors are conceptualized. Many of the fusions and mutations present in these tumors are associated with molecular targets, which have agents in development or already in clinical use. Various agents, including MEK inhibitors, BRAF inhibitors, MTOR inhibitors and, in small subsets of patients NTRK inhibitors, have been used successfully to treat children with recurrent disease, after failure of conventional approaches such as surgery or chemotherapy. The relative benefits of chemotherapy as compared to molecular-targeted therapy for children with newly diagnosed gliomas and neuroglial tumors are under study. Already the combination of an MEK inhibitor and a BRAF inhibitor has been shown superior to conventional chemotherapy (carboplatin and vincristine) in newly diagnosed children with BRAF-V600E mutated low-grade gliomas and neuroglial tumors. However, the long-term effects of such molecular-targeted treatment are unknown. The potential use of molecular-targeted therapy in early treatment has made it mandatory that the molecular make-up of the majority of low-grade glial and glioneuronal tumors is known before initiation of therapy. The primary exception to this rule is in children with neurofibromatosis type 1 who, by definition, have NF1 loss; however, even in this population, gliomas arising in late childhood and adolescence or those not responding to conventional treatment may be candidates for biopsy, especially before entry on molecular-targeted therapy trials.
    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
    根据回顾性队列研究,儿科低度胶质瘤的传播可能发生在约4%-10%的患者中。由于发病率低,对于这些患者的治疗尚无共识.根据MAPK/ERK通路在这些肿瘤中的构成激活,MEK抑制剂如曲美替尼已成功用于复发情况。接受曲美替尼的患者皮肤毒性很常见,通常轻度至中度,但有时严重,需要停止药物,限制肿瘤的疗效。文献中关于减少曲美替尼的剂量是否能够维持疗效的信息不多,同时,降低毒性。这里,我们介绍一个青少年,有严重的皮肤毒性,曲美替尼剂量减少了50%,对肿瘤的疗效持续,而皮肤毒性显着降低。
    Dissemination in pediatric low-grade glioma may occur in about 4%-10% of patients according to retrospective cohort studies. Due to its low incidence, there is no consensus on treatment for these patients. According to the constitutional activation of the MAPK/ERK pathway in these tumors, MEK inhibitors such as trametinib have been used successfully in the relapsed setting. Skin toxicity is frequent in patients receiving trametinib, normally mild to moderate, but sometimes severe, needing to discontinue the drug, limiting the efficacy in the tumor. There is not much information in the literature regarding whether reducing the dose of trametinib is able to maintain efficacy while, at the same time, decreasing toxicity. Here, we present an adolescent, with severe skin toxicity, whose trametinib dose was reduced by 50% and efficacy on the tumor continued while skin toxicity significantly decreased.
    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
    胶质瘤是最常见的成人和儿童原发性脑肿瘤。分子研究已经确定了可以增强诊断并提供生物标志物的特征。具有ATRX和TP53突变的IDH1/2突变定义了弥漫性星形细胞瘤,而具有1p19q缺失的IDH1/2突变定义了少突胶质细胞瘤。受体酪氨酸激酶基因的局灶性扩增,TERT启动子突变,10号染色体和13号染色体缺失,7号染色体三体性是胶质母细胞瘤的特征性特征,可用于诊断。低级别神经胶质瘤中的BRAF基因融合和突变以及高级别神经胶质瘤中的组蛋白H3突变也可用于诊断。
    Gliomas are the most common adult and pediatric primary brain tumors. Molecular studies have identified features that can enhance diagnosis and provide biomarkers. IDH1/2 mutation with ATRX and TP53 mutations defines diffuse astrocytomas, whereas IDH1/2 mutations with 1p19q loss defines oligodendroglioma. Focal amplifications of receptor tyrosine kinase genes, TERT promoter mutation, and loss of chromosomes 10 and 13 with trisomy of chromosome 7 are characteristic features of glioblastoma and can be used for diagnosis. BRAF gene fusions and mutations in low-grade gliomas and histone H3 mutations in high-grade gliomas also can be used for diagnostics.
    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: Review
    背景:发生在中枢神经系统(CNS)的组织细胞肉瘤患者很少见,预后极差。实体瘤中分子诊断方法的增加为中枢神经系统组织细胞肉瘤(CNSHS)的诊断和治疗带来了更多的机会。
    方法:一名9岁女孩因头颈部疼痛入院,还有呕吐。影像学扫描显示前镰状区有明显异常,和组织病理学显示CD68(+)和CD163(+)细胞的存在,导致原发性脑内CNSHS的初步诊断。分子谱分析测试确定了ARHGAP45的一种新变体::BRAF融合在这种情况下,在任何其他肿瘤中均未见报道。患者接受了手术切除肿瘤,需要长期监测。
    结论:BRAF点突变的存在,主要是BRAFp.V600E,已在CNSHS的先前文献中记录。这是前镰状区小儿组织细胞肉瘤的第一例,具有独特的ARHGAP45::BRAF融合。我们的研究结果表明,在CNSHS的诊断中应采用更广泛的分子检测方法,并为患者的治疗开辟了新的可能性。
    BACKGROUND: Patients with histiocytic sarcoma occurring in the central nervous system (CNS) are rare and have a very poor prognosis. The increased use of molecular diagnostic approaches in solid tumors has brought more opportunities for the diagnosis and treatment of central nervous system histiocytic sarcoma (CNSHS).
    METHODS: A 9-year-old girl was admitted to the hospital with pain in her head and neck, as well as vomiting. Imaging scans showed a prominent abnormality in the anterior falciform region, and histopathology revealed the presence of CD68 (+) and CD163 (+) cells, leading to a preliminary diagnosis of primary intracerebral CNSHS. Molecular profiling tests identified a new variant of ARHGAP45::BRAF fusion in this case, which has not been reported in any other tumor. The patient underwent surgical removal of the tumor and will require long-term monitoring.
    CONCLUSIONS: The presence of the BRAF point mutation, predominantly BRAF p.V600E, has been documented in prior literature of CNSHS. This is the first case of pediatric histiocytic sarcoma in the anterior falciform region who has a unique ARHGAP45::BRAF fusion. The findings of our study indicate that a broader range of molecular assays should be employed in the diagnosis of CNSHS and opens up new possibilities for the treatment of the patient.
    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:我们旨在基于从多参数MRI提取的影像组学特征,开发机器学习模型,用于预测儿童低级别神经胶质瘤(PLGG)的分子亚群(低危组和中/高危组)和分子标记(KIAA1549-BRAF融合)。
    方法:本回顾性研究包括61例PLGG患者,根据分子亚组或分子标记以2:1的比例分为训练集和内部验证集。将患者分为低风险和中/高风险组,BRAF融合阳性和阴性组,分别。我们从多参数MRI中提取了5929个放射学特征。此后,我们删除了多余的功能,在训练集上训练随机森林模型,用于预测分子亚群或分子标记,并在内部验证集上验证了它们的性能。通过3折交叉验证验证了预测模型的性能。
    结果:我们使用4个相关特征构建了区分低风险PLGs和中/高风险PLGs的分类模型,内部验证集的AUC为0.833,准确率为76.2%。在使用4个相关特征预测KIAA1549-BRAF融合的预测模型中,内部验证集的AUC为0.818,准确率为81.0%.
    结论:目前的研究表明,MRI影像组学能够以令人满意的灵敏度预测PLGG和KIAA1549-BRAF融合的分子亚群。
    背景:本研究在clinicaltrials.gov(NCT04217018)进行了回顾性注册。
    BACKGROUND: We aimed to develop machine learning models for prediction of molecular subgroups (low-risk group and intermediate/high-risk group) and molecular marker (KIAA1549-BRAF fusion) of pediatric low-grade gliomas (PLGGs) based on radiomic features extracted from multiparametric MRI.
    METHODS: 61 patients with PLGGs were included in this retrospective study, which were divided into a training set and an internal validation set at a ratio of 2:1 based on the molecular subgroups or the molecular marker. The patients were classified into low-risk and intermediate/high-risk groups, BRAF fusion positive and negative groups, respectively. We extracted 5929 radiomic features from multiparametric MRI. Thereafter, we removed redundant features, trained random forest models on the training set for predicting the molecular subgroups or the molecular marker, and validated their performance on the internal validation set. The performance of the prediction model was verified by 3-fold cross-validation.
    RESULTS: We constructed the classification model differentiating low-risk PLGGs from intermediate/high-risk PLGGs using 4 relevant features, with an AUC of 0.833 and an accuracy of 76.2% in the internal validation set. In the prediction model for predicting KIAA1549-BRAF fusion using 4 relevant features, an AUC of 0.818 and an accuracy of 81.0% were achieved in the internal validation set.
    CONCLUSIONS: The current study demonstrates that MRI radiomics is able to predict molecular subgroups of PLGGs and KIAA1549-BRAF fusion with satisfying sensitivity.
    BACKGROUND: This study was retrospectively registered at clinicaltrials.gov (NCT04217018).
    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
    毛细胞星形细胞瘤(PA),中枢神经系统(CNS)世界卫生组织1级肿瘤,主要见于5-19岁的儿童或年轻人。手术切除通常提供优异的结果,但残余肿瘤可能仍然存在。这种低度肿瘤因其经典的放射学和形态学特征而广为人知;然而,下一代测序(NGS)的应用揭示了一些独特的分子发现.在这种低度肿瘤中发现的遗传异常中,越来越多的证据表明BRAF改变,尤其是BRAF融合,在PA肿瘤发生中起重要作用。在PAs中鉴定的几个融合伴侣基因中,KIAA1549-BRAF融合是最常见的可检测遗传改变,尤其是在小脑PA中。这里,我们报告了一个年轻的成年患者,右侧后颅窝小脑和小脑桥脑角区域肿块与PA一致。值得注意的是,NGS检测到一种新的GNAI3-BRAF融合,其产生含有BRAF的激酶结构域的框内融合蛋白。这一发现扩展了BRAF融合在PAs肿瘤发生中的知识,为诊断提供了额外的分子特征,和未来治疗的目标。
    Pilocytic astrocytoma (PA), a central nervous system (CNS) World Health Organization grade 1 tumor, is mainly seen in children or young adults aged 5-19. Surgical resection often provides excellent outcomes, but residual tumors may still remain. This low-grade tumor is well recognized for its classic radiological and morphological features; however, some unique molecular findings have been unveiled by the application of next-generation sequencing (NGS). Among the genetic abnormalities identified in this low-grade tumor, increasing evidence indicates that BRAF alterations, especially BRAF fusions, play an essential role in PA tumorigenesis. Among the several fusion partner genes identified in PAs, KIAA1549-BRAF fusion is notably the most common detectable genetic alteration, especially in the cerebellar PAs. Here, we report a case of a young adult patient with a large, right-sided posterior fossa cerebellar and cerebellopontine angle region mass consistent with a PA. Of note, NGS detected a novel GNAI3-BRAF fusion, which results in an in-frame fusion protein containing the kinase domain of BRAF. This finding expands the knowledge of BRAF fusions in the tumorigenesis of PAs, provides an additional molecular signature for diagnosis, and a target for future 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
    未经证实:非小细胞肺癌(NSCLC)是世界上最常见的恶性肿瘤之一。Osimertinib是第三代表皮生长因子受体酪氨酸激酶抑制剂(EGFR-TKI),可有效靶向敏感的表皮生长因子受体突变和外显子20T790M。尽管最初取得了令人印象深刻的结果,获得性抗性(AR)发展迅速,通常在9-13个月内,和抵抗的机制还没有完全理解。在过去的几年里,EGFR-TKI和程序性细胞死亡配体1(PD-L1)抑制剂已被广泛用于治疗晚期肺腺癌患者。
    UASSIGNED:在此,我们报告了一名中年女性,根据病理诊断患有肺腺癌。通过下一代测序(NGS)检测到表皮生长因子受体外显子19的缺失。在患者接受了一系列治疗后,包括奥希替尼,鉴定了BTN2A1-BRAF融合。在通过免疫组织化学(IHC)评估PD-L1表达后,患者改用duvalizumab,PD-L1抑制剂,但没有观察到显著的改善。进行NGS和IHC测定以分析在治疗期间获得的活检和血液样品。
    未经批准:该病例证实,获得BTN2A1-BRAF融合可能是NSCLC中AR与奥希替尼的一种机制。与IHC中肿瘤组织中PD-L1表达的程度无关,具有敏感表皮生长因子受体突变的患者从PD-L1抑制剂获得的益处最小。我们的病例为治疗该患者人群提供了新的思路。
    UNASSIGNED: Non-small cell lung cancer (NSCLC) is one of the most common malignancies in the world. Osimertinib is a third-generation epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) indicated for NSCLC that effectively targets sensitive epidermal growth factor receptor mutation and exon20 T790M. Despite initially impressive outcomes, acquired resistance (AR) develops rapidly, typically within 9-13 months, and the mechanisms of resistance are not fully understood. Over the past years, EGFR-TKI and programmed cell death-ligand 1 (PD-L1) inhibitors have been widely used to treat for patients with advanced lung adenocarcinoma.
    UNASSIGNED: Herein we report a middle-aged female who suffered from lung adenocarcinoma based on the pathological diagnosis. Epidermal growth factor receptor exon 19 deletion was detected by next-generation sequencing (NGS). After the patient underwent a series of treatments, including osimertinib, BTN2A1-BRAF fusion was identified. After assessing PD-L1 expression by immunohistochemistry (IHC), the patient was switched to duvalizumab, a PD-L1 inhibitor, but no significant improvements were observed. NGS and IHC assays were conducted to analyze the biopsy and blood samples obtained during treatment.
    UNASSIGNED: This case substantiates that the acquisition of BTN2A1-BRAF fusion potentially serves as a mechanism of AR to osimertinib in NSCLC. Patients with sensitive epidermal growth factor receptor mutation derive minimal benefit from PD-L1 inhibitors irrespective of the degree of PD-L1 expression in the tumor tissue in IHC. Our case provides a new train of thought for treating 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: Case Reports
    BRAF基因已被确定为致癌驱动因子和各种恶性肿瘤的潜在靶标。BRAF融合是罕见频率的BRAF改变的一种亚型。这里,我们首先报道了1例既往接受SND1-BRAF从头融合治疗的晚期肺腺癌患者,该患者对MAK抑制剂曲美替尼实现部分应答.我们还提供了关于BRAF融合的靶向治疗的文献综述。
    BRAF gene has been identified as an oncogenic driver and a potential target in various malignancies. BRAF fusions are one subtype of BRAF alterations with a rare frequency. Here, we first report a previously treated advanced lung adenocarcinoma patient with de novo SND1-BRAF fusion who achieves partial response to the MAK inhibitor trametinib. We also provide a literature review on targeted therapies for BRAF fusions.
    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
    BRAF融合是非小细胞肺癌(NSCLC)中罕见的驱动癌基因。类似于BRAFV600E突变,它还可以激活MAPK信号通路。有一些病例报告表明对BRAF抑制剂的潜在反应及其作为从头驱动突变的重要作用。此外,同时发生的MET扩增已被定义为表皮生长因子受体(EGFR)突变型NSCLC患者的不良预后因素.目前,目前正在进行的临床试验研究MET扩增作为EGFR突变型NSCLC和奥希替尼获得性耐药患者的治疗靶点,这意味着MET扩增也具有治疗意义。然而,之前未在BRAF融合患者中对同时发生的MET扩增进行过研究.一名67岁的男子被诊断为转移性低分化腺癌。他接受了pembrolizumab和化疗联合的一线治疗,因为基因组检测显示野生型EGFR,免疫组织化学染色显示ALK和ROS1阴性。随着疾病进展,下一代测序显示KIAA1549-BRAF融合和MET扩增同时发生.随后的Dabrafenib,曲美替尼,卡马替尼联合治疗效果显著。针对共同发生的驱动突变的联合治疗是NSCLC患者的潜在有效治疗。仍需要进一步的前瞻性研究来研究共同发生的驱动突变的作用和相关的治疗策略。
    BRAF fusions are rare driver oncogenes in non-small cell lung cancer (NSCLC). Similar with BRAF V600E mutation, it could also activate the MAPK signaling pathway. There are a few case reports which had indicated the potential response to BRAF inhibitors and its important role as de novo driver mutation. In addition, the co-occurring MET amplification has been defined as a poor prognostic factor in patients with epidermal growth factor receptor (EGFR) mutant NSCLC. Currently, there are ongoing clinical trials which investigate the MET amplification as a therapeutic target in patients with EGFR mutant NSCLC and acquired resistance to osimertinib, which imply that the MET amplification also had a therapeutic significance. However, the co-occurring MET amplification had not been studied in patients with BRAF fusion before. A 67-year-old man was diagnosed with metastatic poorly-differentiated adenocarcinoma. He received first-line therapy with the combination of pembrolizumab and chemotherapy because the genomic test revealed wild-type EGFR, and negativity of ALK and ROS1 by immunohistochemical stain. Upon disease progression, the next-generation sequencing revealed co-occurring KIAA1549-BRAF fusion and MET amplification. Subsequent dabrafenib, trametinib, and capmatinib combination therapy showed a remarkable treatment effect. The combination therapy targeting the co-occurring driver mutations is a potential effective treatment for NSCLC patients. Further prospective study is still warranted to investigate the role of co-occurring driver mutations and the relevant treatment strategy.
    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
    Osimertinib has become a standard of care in the first-line treatment of advanced-stage non-small-cell lung cancer (NSCLC) harboring exon 19 and 21 activating mutations in the EGFR gene. Nevertheless, the 18.9-month median progression-free survival emphasizes the fact that resistance to osimertinib therapy is inevitable. Acquired resistance mechanisms to osimertinib in EGFR-driven NSCLC include MET amplification, EGFR C797S mutation, neuroendocrine differentiation, small-cell lung carcinoma histologic transformation, PD-L1 and KRAS amplifications and ESR1-AKAP12 and MKRN1-BRAF translocations, as well as BRAF V600 mutation. This last one represents 3% of the acquired resistance mechanisms to osimertinib. In this review, we discuss the rationale for EGFR/BRAF/MEK co-inhibition in the light of a clinical case of EGFR-mutant NSCLC developing a BRAF V600 mutation as an acquired resistance mechanism to osimertinib and responding to the association of osimertinib plus dabrafenib and trametinib. Additionally, we discuss the acquired resistance mechanisms to osimertinib plus dabrafenib and trametinib combination in that context.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号