NTRK fusions

NTRK 融合
  • 文章类型: Journal Article
    神经营养受体激酶(NTRK)融合是多种儿童和成人实体瘤的可行致癌驱动因素,原肌球蛋白受体激酶(TRK)被认为是具有这些融合的“泛癌症”的有吸引力的治疗靶标。目前,已经开发了两代TRK抑制剂。具有代表性的第二代抑制剂selitrectinib和repotrectinib旨在克服由溶剂前沿和看门人靶突变引起的第一代抑制剂larotrectinib或entrectinib的临床获得性耐药性。然而,xDFG(TRKAG667C/A/S,同源TRKCG696C/A/S)和一些双重突变仍然赋予对selitrectinib和repotrectinib的抗性,克服这些耐药性代表了一个主要的未满足的临床需求。在这次审查中,我们总结了第一代和第二代TRK抑制剂的获得性耐药机制,并首次提出了新兴的选择性II型TRK抑制剂来克服xDFG突变介导的耐药性。此外,我们总结了我们对这一领域新挑战和未来方向的看法。
    Neurotrophic receptor kinase (NTRK) fusions are actionable oncogenic drivers of multiple pediatric and adult solid tumors, and tropomyosin receptor kinase (TRK) has been considered as an attractive therapeutic target for \"pan-cancer\" harboring these fusions. Currently, two generations TRK inhibitors have been developed. The representative second-generation inhibitors selitrectinib and repotrectinib were designed to overcome clinic acquired resistance of the first-generation inhibitors larotrectinib or entrectinib resulted from solvent-front and gatekeeper on-target mutations. However, xDFG (TRKAG667C/A/S, homologous TRKCG696C/A/S) and some double mutations still confer resistance to selitrectinib and repotrectinib, and overcoming these resistances represents a major unmet clinical need. In this review, we summarize the acquired resistance mechanism of the first- and second-generation TRK inhibitors, and firstly put forward the emerging selective type II TRK inhibitors to overcome xDFG mutations mediated resistance. Additionally, we concluded our perspectives on new challenges and future directions in this field.
    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
    目的:NTRK融合导致组成型活性致癌TRK蛋白,约占非小细胞肺癌(NSCLC)病例的0.2%。大约40%的晚期NSCLC患者发生CNS转移;因此,需要具有颅内(IC)疗效的治疗。在对三个I/II期研究的综合分析中(ALKA-372-001:EudraCT2012-000148-88;STARTRK-1:NCT02097810;STARTRK-2:NCT02568267),恩替尼,一个强大的,中枢神经系统活性,TRK抑制剂,NTRK融合阳性(fp)NSCLC患者的疗效(客观缓解率[ORR]:64.5%;2021年8月2日数据截止)。我们提供了该队列的最新数据。
    方法:符合条件的患者为≥18岁的局部晚期/转移性患者,NTRK-fpNSCLC随访≥12个月。在第4周和此后每8周通过根据RECISTv1.1的盲法独立中央审查(BICR)评估肿瘤应答。共同主要终点:ORR;反应持续时间(DoR)。次要终点包括无进展生存期(PFS);总生存期(OS);IC疗效;安全性。入学截止日期:2021年7月2日;数据截止日期:2022年8月2日。
    结果:可评价疗效的人群包括51例NTRK-fpNSCLC患者。中位年龄为60.0岁(范围22-88);20例患者(39.2%)有研究者评估的基线CNS转移。中位生存期随访为26.3个月(95%CI21.0-34.1)。ORR为62.7%(95%CI48.1-75.9),有6个完整和26个部分响应。平均DoR和PFS分别为27.3个月(95%CI19.9-30.9)和28.0个月(95%CI15.7-30.4),分别。中位OS为41.5个月。在BICR评估基线中枢神经系统转移的患者中,IC-ORR为64.3%(n=9/14;95%CI35.1-87.2),包括七个完整的响应者,IC-DoR为55.7个月。在安全性可评估的人群中(n=55),大多数治疗相关不良事件为1/2级;未报告治疗相关死亡.
    结论:Entrectinib在NTRK-fpNSCLC患者中继续表现出深度和持久的全身和IC反应。
    NTRK fusions result in constitutively active oncogenic TRK proteins responsible for ∼ 0.2 % of non-small cell lung cancer (NSCLC) cases. Approximately 40 % of patients with advanced NSCLC develop CNS metastases; therefore, treatments with intracranial (IC) efficacy are needed. In an integrated analysis of three phase I/II studies (ALKA-372-001: EudraCT 2012-000148-88; STARTRK-1: NCT02097810; STARTRK-2: NCT02568267), entrectinib, a potent, CNS-active, TRK inhibitor, demonstrated efficacy in patients with NTRK fusion-positive (fp) NSCLC (objective response rate [ORR]: 64.5 %; 2 August 2021 data cut-off). We present updated data for this cohort.
    Eligible patients were ≥ 18 years with locally advanced/metastatic, NTRK-fp NSCLC with ≥ 12 months of follow-up. Tumor responses were assessed by blinded independent central review (BICR) per RECIST v1.1 at Week 4 and every eight weeks thereafter. Co-primary endpoints: ORR; duration of response (DoR). Secondary endpoints included progression-free survival (PFS); overall survival (OS); IC efficacy; safety. Enrolment cut-off: 2 July 2021; data cut-off: 2 August 2022.
    The efficacy-evaluable population included 51 patients with NTRK-fp NSCLC. Median age was 60.0 years (range 22-88); 20 patients (39.2 %) had investigator-assessed baseline CNS metastases. Median survival follow-up was 26.3 months (95 % CI 21.0-34.1). ORR was 62.7 % (95 % CI 48.1-75.9), with six complete and 26 partial responses. Median DoR and PFS were 27.3 months (95 % CI 19.9-30.9) and 28.0 months (95 % CI 15.7-30.4), respectively. Median OS was 41.5 months. In patients with BICR-assessed baseline CNS metastases, IC-ORR was 64.3 % (n = 9/14; 95 % CI 35.1-87.2), including seven complete responders, and IC-DoR was 55.7 months. In the safety-evaluable population (n = 55), most treatment-related adverse events were grade 1/2; no treatment-related deaths were reported.
    Entrectinib has continued to demonstrate deep and durable systemic and IC responses in patients with NTRK-fp NSCLC.
    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:肿瘤基因分型对于优化晚期分化型甲状腺癌(DTC)患者的临床管理变得至关重要;然而,其在临床实践中的实施仍未定义。我们在此报告了我们通过下一代测序方法进行分子高级DTC测试的单中心经验,为了更好地定义肿瘤基因分型的方式和时间可以帮助临床决策。
    方法:我们回顾性收集了2008年至2022年在IRCSSSant'Orsola-Malpighi医院接受分子谱分析的所有晚期DTC成年患者的数据。遗传改变与放射性碘化物难治性(RAI-R)相关,RAI摄取/疾病状态,以及RAI抗性(TTRR)发展的时间。
    结果:发现RAI-R发育与遗传改变之间存在显着相关性(P=0.0001)。大约48.7%的RAI-R病例对TERT/TP53突变呈阳性(作为单一事件和与其他驱动基因改变的合并,比如BRAF突变,RAS突变,或基因融合),而绝大多数RAI敏感病例携带基因融合(41.9%)或野生型(WT;41.9%)。RAI摄取/疾病状态和到达TTRR的时间与遗传改变显著相关(P=0.0001)。特别是,以TERT/TP53突变作为单一事件或合并的DTC显示较短的中位TTRR为35.4个月(范围15.0-55.8个月),与其他分子亚组相比。在Cox多变量分析后,TERT/TP53突变作为单个事件或合并与RAI-R独立相关(风险比4.14,95%CI1.51-11.32;P=0.006)。
    结论:常规检测基因改变应作为临床检查的一部分,用于识别更具侵袭性的肿瘤的子集和具有可操作基因融合的肿瘤的子集,从而确保所有晚期DTC患者的适当管理。
    BACKGROUND: Tumor genotyping is becoming crucial to optimize the clinical management of patients with advanced differentiated thyroid cancer (DTC); however, its implementation in clinical practice remains undefined. We herein report our single-center experience on molecular advanced DTC testing by next-generation sequencing approach, to better define how and when tumor genotyping can assist clinical decision making.
    METHODS: We retrospectively collected data on all adult patients with advanced DTC who received molecular profiling at the IRCSS Sant\'Orsola-Malpighi Hospital from 2008 to 2022. The genetic alterations were correlated with radioactive iodide refractory (RAI-R), RAI uptake/disease status, and time to RAI resistance (TTRR) development.
    RESULTS: A significant correlation was found between RAI-R development and genetic alterations (P = 0.0001). About 48.7% of RAI-R cases were positive for TERT/TP53 mutations (as both a single event and comutations with other driver gene alterations, such as BRAF mutations, RAS mutations, or gene fusions), while the great majority of RAI-sensitive cases carried gene fusions (41.9%) or were wild type (WT; 41.9%). RAI uptake/disease status and time to TTRR were significantly associated with genetic alterations (P = 0.0001). In particular, DTC with TERT/TP53 mutations as a single event or as comutations displayed a shorter median TTRR of 35.4 months (range 15.0-55.8 months), in comparison to the other molecular subgroups. TERT/TP53 mutations as a single event or as comutations remained independently associated with RAI-R after Cox multivariate analysis (hazard ratio 4.14, 95% CI 1.51-11.32; P = 0.006).
    CONCLUSIONS: Routine testing for genetic alterations should be included as part of the clinical workup, for identifying both the subset of more aggressive tumors and the subset of tumors harboring actionable gene fusions, thus ensuring the appropriate management for all patients with advanced DTC.
    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
    三阴性乳腺癌(TNBC)通常是一种侵袭性疾病,预后差,治疗选择有限。神经营养酪氨酸受体激酶(NTRK)基因融合是由食品和药物管理局(FDA)批准的癌症类型不可知的新兴生物标志物。美国,用于选择患者进行靶向治疗。我们研究的主要目的是调查NTRK像差的频率,即融合,基因拷贝数增加,和放大,在一系列TNBC中使用不同的方法。使用pan-TRK免疫组织化学(IHC)分析了总共83个TNBC,荧光原位杂交(FISH),实时聚合酶链反应(RT-PCR),和基于RNA的下一代测序(NGS)。83例,16显示pan-TRK阳性,尽管没有NTRK融合。的确,FISH显示4例携带由一个融合信号和一个/多个单一绿色信号组成的非典型NTRK1模式,但通过NGS和RT-PCR检测,所有病例均为融合阴性。此外,FISH分析显示6例NTRK1扩增,一个具有NTRK2拷贝数增益的情况,5例NTRK3拷贝数增加,泛TRKIHC均为阴性。我们的数据表明,IHC对融合体的检测具有很高的假阳性率,并且必须进行分子检测;如果IHC对NTRK阴性,则无需进行其他分子检测。总之,NTRK基因不参与TNBC的融合,但是拷贝数增加和扩增都是频繁的事件,提示其他NTRK像差可能的预测作用。
    Triple-negative breast cancer (TNBC) is usually an aggressive disease with a poor prognosis and limited treatment options. The neurotrophic tyrosine receptor kinase (NTRK) gene fusions are cancer type-agnostic emerging biomarkers approved by the Food and Drug Administration (FDA), USA, for the selection of patients for targeted therapy. The main aim of our study was to investigate the frequency of NTRK aberrations, i.e. fusions, gene copy number gain, and amplification, in a series of TNBC using different methods. A total of 83 TNBCs were analyzed using pan-TRK immunohistochemistry (IHC), fluorescence in situ hybridization (FISH), real-time polymerase chain reaction (RT-PCR), and RNA-based next-generation sequencing (NGS). Of 83 cases, 16 showed pan-TRK positivity although no cases had NTRK-fusions. Indeed, FISH showed four cases carrying an atypical NTRK1 pattern consisting of one fusion signal and one/more single green signals, but all cases were negative for fusion by NGS and RT-PCR testing. In addition, FISH analysis showed six cases with NTRK1 amplification, one case with NTRK2 copy number gain, and five cases with NTRK3 copy number gain, all negative for pan-TRK IHC. Our data demonstrate that IHC has a high false-positive rate for the detection of fusions and molecular testing is mandatory; there is no need to perform additional molecular tests in cases negativity for NTRK by IHC. In conclusion, the NTRK genes are not involved in fusions in TNBC, but both copy number gain and amplification are frequent events, suggesting a possible predictive role for other NTRK aberrations.
    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
    原肌球蛋白受体激酶(TRK)抑制剂已被批准用于携带NTRK融合的转移性实体瘤,但是NTRK融合的检测具有挑战性。国际指南建议泛TRK免疫组织化学(IHC)筛查,然后在NTRK融合患病率低的肿瘤类型中进行下一代测序(NGS)。包括转移性结直肠癌(mCRC)。基于RNA的NGS是优选的,但是很贵,耗时,从FFPE组织中提取高质量的RNA具有挑战性。日常临床实践中的替代方案是必要的。我们评估了RNA-NGS的诊断性能,FFPE靶向基因座捕获(FFPE-TLC),荧光原位杂交(FISH),和5'/3'不平衡定量RT-PCR(qRT-PCR)的IHC筛查后的268例微卫星不稳定性高mCRC患者,NTRK融合最普遍的亚组(1-5%)。在审查所有测定结果后确定一致结果。在16个IHC阳性肿瘤中,检测到10个NTRK融合。在33个IHC阴性样本中,没有发现其他转录的NTRK融合体,强调了IHC的高灵敏度。RNA-NGS的敏感性,FFPE-TLC,FISH,qRT-PCR为90%,90%,78%,100%,分别。所有测定的特异性为100%。鲁棒性,定义为在第一次运行中提供可解释结果的样本百分比,FFPE-TLC为100%,RNA-NGS(85%)更有限,鱼(70%),和qRT-PCR(70%)。总的来说,我们不推荐FISH用于mCRC中NTRK融合的检测,因为其灵敏度低,鲁棒性有限.我们得出结论,RNA-NGS,FFPE-TLC,qRT-PCR是NTRK融合检测的合适方法,用泛TRKIHC富集后,在常规临床实践中。
    Tropomyosin receptor kinase (TRK) inhibitors have been approved for metastatic solid tumors harboring NTRK fusions, but the detection of NTRK fusions is challenging. International guidelines recommend pan-TRK immunohistochemistry (IHC) screening followed by next generation sequencing (NGS) in tumor types with low prevalence of NTRK fusions, including metastatic colorectal cancer (mCRC). RNA-based NGS is preferred, but is expensive, time-consuming, and extracting good-quality RNA from FFPE tissue is challenging. Alternatives in daily clinical practice are warranted. We assessed the diagnostic performance of RNA-NGS, FFPE-targeted locus capture (FFPE-TLC), fluorescence in situ hybridization (FISH), and the 5\'/3\' imbalance quantitative RT-PCR (qRT-PCR) after IHC screening in 268 patients with microsatellite-instability-high mCRC, the subgroup in which NTRK fusions are most prevalent (1-5%). A consensus result was determined after review of all assay results. In 16 IHC positive tumors, 10 NTRK fusions were detected. In 33 IHC negative samples, no additional transcribed NTRK fusions were found, underscoring the high sensitivity of IHC. Sensitivity of RNA-NGS, FFPE-TLC, FISH, and qRT-PCR was 90%, 90%, 78%, and 100%, respectively. Specificity was 100% for all assays. Robustness, defined as the percentage of samples that provided an interpretable result in the first run, was 100% for FFPE-TLC, yet more limited for RNA-NGS (85%), FISH (70%), and qRT-PCR (70%). Overall, we do not recommend FISH for the detection of NTRK fusions in mCRC due to its low sensitivity and limited robustness. We conclude that RNA-NGS, FFPE-TLC, and qRT-PCR are appropriate assays for NTRK fusion detection, after enrichment with pan-TRK IHC, in routine clinical practice.
    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
    胰腺导管腺癌(PDAC)预后差,美国只有10%的患者从诊断之日起存活5年。直到最近,根据患者或肿瘤特征,晚期PDAC的治疗差异不大.然而,最近的突破已经确定了受益于新的患者亚组,生物标志物驱动的疗法。我们回顾了PARP抑制剂和其他生物标志物导向疗法的数据和作用。包括NTRK融合患者,NRG1融合,错配修复缺陷,和KRASp.G12C突变。
    Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis, with a mere ∼10% of patients in the United States surviving 5 years from the time of diagnosis. Until recently, the treatment for advanced PDAC differed little based on patient or tumor characteristics. However, recent breakthroughs have identified subgroups of patients who benefit from novel, biomarker-driven therapies. We review the data and role for PARP inhibitors and for other biomarker-directed therapies, including for patients with NTRK fusions, NRG1 fusions, mismatch repair deficiency, and KRAS p.G12C mutations.
    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
    背景:NTRK(神经营养性酪氨酸受体激酶)重排的梭形细胞肿瘤是一组新的肿瘤,包括在新的第5版世界卫生组织(WHO)软组织和骨肉瘤分类中。这些肿瘤的特征在于NTRK基因融合,并显示广谱的组织学和临床行为。一些靶向疗法最近已被批准用于携带NTRK融合的肿瘤。包括STS。
    方法:一名26岁男性,预处理的NTRK重排梭形细胞肿瘤和肝脏,肺和骨转移用拉罗列替尼连续28天治疗,剂量为100毫克,每天两次。治疗7天后进行的18FDG-PET/CT扫描显示内脏和骨病变中的肿瘤缩小。没有药物相关的毒性。随后的评估证实了疾病部位的肿瘤持续消退。患者情况良好,继续治疗。
    结论:使用第一代TRK抑制剂治疗的罕见TPM4(外显子7)-NTRK1(外显子12)基因融合肿瘤患者的临床和放射学反应可能有助于更好地了解这种新STS实体的生物学特性,并有助于改善患者管理。
    BACKGROUND: NTRK (neurotrophic tyrosine receptor kinase)-rearranged spindle cell neoplasms are a new group of tumors included in the new 5th edition of the World Health Organization (WHO) classification of soft Tissue and Bone Sarcomas. These tumors are characterized by NTRK gene fusions and show a wide spectrum of histologies and clinical behavior. Several targeted therapies have recently been approved for tumors harboring NTRK fusions, including STS.
    METHODS: A 26-year-old male with advanced, pretreated NTRK rearranged spindle cell neoplasm and liver, lung and bone metastases was treated with larotrectinib on a continuous 28-day schedule, at a dose of 100 mg twice daily. An 18FDG-PET/CT scan performed after 7 days of treatment showed tumor shrinkage in both visceral and bone lesions. There was no drug-related toxicity. Subsequent evaluations confirmed continued tumor regression in disease sites. The patient is well and continues treatment.
    CONCLUSIONS: The clinical and radiological response of our patient with an uncommon TPM4 (exon 7)-NTRK1 (exon 12) gene fusion tumor treated with a first-generation TRK inhibitor could contribute to a better understanding of the biology of this new STS entity and help to improve patient management.
    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
    使用pan-TRKIHC等方法可以在肿瘤类型中检测到可靶向的NTRK基因融合体,DNA或RNANGS测试,或者鱼。实施NTRK融合的临床测试的挑战可能由于NTRK融合在肿瘤中的流行范围而出现。组织中TRK表达的内源性水平,以及大量潜在的融合伙伴。在这项研究中,我们检查了我们评估驱动突变阴性肺的经验,尿路上皮或胆管癌病例,除了阳性的病例,模棱两可,或通过pan-TRKIHC对NTRK融合体的弱染色。其中63/127(49.6%)为泛TRK免疫组化阳性,其中71.4%表现为微弱或局灶性染色,可能是由于生理或非特异性TRK表达。在这127个案例中,如通过RNA融合组测试所证实的,图4中所示的具有NTRK融合体(在来自同一患者的两个单独的样品中观察到1个融合体)。Pan-TRK免疫组化阳性1例TPM3-NTRK1融合,1例GOLGA4-NTRK3融合不一致,在2个ADAM19-NTRK3融合样品中呈阴性。我们的研究结果表明,我们能够成功地鉴定出导致靶向治疗的NTRK融合。然而,我们的结果表明pan-TRKIHC对NTRK3融合的敏感性有限,并且在具有生理性或非特异性TRK表达的肿瘤中pan-TRKIHC的特异性降低,结果假阳性样品需要通过基于RNA的NGS进行验证性测试。
    Targetable NTRK gene fusions can be detected across tumor types using methodologies such as pan-TRK IHC, DNA or RNA NGS testing, or FISH. Challenges for implementation of clinical testing for NTRK fusions may arise due to the range in NTRK fusion prevalence across tumors, endogenous levels of TRK expression in tissues, and the large number of potential fusion partners. In this study, we examined our experience evaluating driver mutation negative lung, urothelial or cholangiocarcinoma cases, in addition to cases with positive, equivocal, or weak staining by pan-TRK IHC for NTRK fusions. 63/127 (49.6%) of these cases were positive for pan-TRK IHC, of which 71.4% showed weak or focal staining, potentially due to physiologic or non-specific TRK expression. Of these 127 cases, 4 harbored a NTRK fusion (1 fusion was seen in two separate samples from the same patient) as confirmed by RNA fusion panel testing. Pan-TRK IHC was positive in 1 case with TPM3-NTRK1 fusion, equivocal in 1 case with GOLGA4-NTRK3 fusion, and negative in 2 samples with ADAM19-NTRK3 fusion. Our findings show that we were able to successfully identify NTRK fusions that resulted in targeted therapy. However, our results suggest limited sensitivity of pan-TRK IHC for NTRK3 fusions, and that the reduced specificity for pan-TRK IHC in tumors with physiologic or non-specific TRK expression, results in false positive samples that require confirmatory testing by RNA based NGS.
    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
    该研究的目的是确定NTRK融合阳性或RET融合/突变阳性的甲状腺癌患者,谁可以受益于神经营养性酪氨酸激酶受体(NTRK)或受体酪氨酸激酶(RET)抑制剂。
    在卡尔加里前瞻性甲状腺癌数据库中确定了患者(N=482)。使用临床可用的MassARRAY®BRAF测试对患者进行预筛选,结肠面板,黑色素瘤小组,或ThyroSPEC™。用Oncomine™综合测定v3(OCAv3)筛选突变阴性肿瘤的NTRK融合和RET融合/突变。
    共有86名患者被纳入2个独立分析中的1个。分析A包括42例放射性碘(RAI)耐药的远处转移患者。预筛查后,20例BRAF和RAS突变阴性患者接受了OCAv3筛查,结果检测到4例NTRKfusions患者和4例RET融合患者(8/20,占分析患者的40%)。分析B包括44例患者,42与美国甲状腺协会(ATA)高和中间复发风险和2与甲状腺髓样癌。在筛查前,1例NTRK融合患者,1例RET融合患者,并鉴定出30例BRAF突变患者.其余9例患者接受OCAv3筛查,结果检测到1例NTRK融合患者和1例RET融合患者(4/11,占分析患者的36%)。
    我们的研究结果表明,在有RAI耐药远处转移和ATA高或中等复发风险的甲状腺癌患者中,NTRK融合和RETfusion的发生率更高。这凸显了早期筛查对NTRK或RET抑制剂干预的重要性。
    The aim of the study was to identify patients with NTRK fusion-positive or RET fusion/mutation-positive thyroid cancers, who could benefit from neurotrophic tyrosine kinase receptor (NTRK) or receptor tyrosine kinase (RET) inhibitors.
    Patients were identified in the Calgary prospective thyroid cancer database (N= 482). Patients were \'pre-screened\' with clinically available MassARRAY® BRAF test, Colon Panel, Melanoma Panel, or ThyroSPEC™. Mutation-negative tumors were \'screened\' for NTRK fusions and RET fusions/mutations with the Oncomine™ Comprehensive Assay v3 (OCAv3).
    A total of 86 patients were included in 1 of 2 separate analyses. Analysis A included 42 patients with radioactive iodine (RAI)-resistant distant metastases. After pre-screening, 20 BRAF and RAS mutation-negative patients underwent OCAv3 screening, resulting in the detection of 4 patients with NTRKfusions and 4 patients with RET fusions (8/20, 40% of analyzed patients). Analysis B included 44 patients, 42 with American Thyroid Association (ATA) high and intermediate risk of recurrence and 2 with medullary thyroid carcinoma. During pre-screening, 1 patient with an NTRK fusion, 1 patient with a RET fusion, and 30 patients with BRAF mutations were identified. The remaining 9 patients received OCAv3 screening, resulting in detection of 1 patient with an NTRKfusion and 1 with a RET fusion (4/11, 36% of analyzed patients).
    Our findings indicate a higher rate of NTRK fusions and RETfusions in patients with thyroid cancer with RAI-resistant distant metastases and ATA high or intermediate risk of recurrence. This highlights the importance of early screening to enable intervention with a NTRK or RET inhibitor.
    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
    涉及TRK蛋白酪氨酸激酶的融合是儿童和成人多种肿瘤的致癌驱动因素,非小细胞肺癌的患病率为0.2%。由于NTRK内含子长度等结构特征,诊断可能具有挑战性。但是下一代测序(NGS),包括基于RNA的NGS,增加检测。第一代TRK抑制剂,拉罗列替尼和恩特列替尼,已经以肿瘤无关的方式在TRK融合阳性癌症中表现出临床意义的抗肿瘤活性,应被视为TRK融合阳性肺癌的一线治疗选择。此外,第一代TRK抑制剂耐受性良好.应该小心,然而,为了监测目标不良事件,如头晕,体重增加,感觉异常,和戒断疼痛。可能发生介导TRK抑制剂抗性的中靶和脱靶机制。下一代TRK抑制剂,比如selitrectinib,repotrectinib,和Taletrectinib,可用于正在进行的临床试验,并解决目标耐药问题。本文将重点介绍NTRK融合和TRK定向靶向治疗在肺癌中的应用。
    Fusions involving TRK protein tyrosine kinases are oncogenic drivers in a variety of tumors in children and adults, with a prevalence of ∼0.2% in non-small cell lung cancer. Diagnosis can be challenging due to structural features such as NTRK intron length, but next-generation sequencing (NGS), including RNA-based NGS, increases detection. The first-generation TRK inhibitors, larotrectinib and entrectinib, have demonstrated clinically meaningful antitumor activity in TRK fusion-positive cancers in a tumor-agnostic fashion and should be considered first-line therapeutic options for TRK fusion-positive lung cancers. Furthermore, the first-generation TRK inhibitors are well tolerated. Care should be taken, however, to monitor on-target adverse events, such as dizziness, weight gain, paresthesias, and withdrawal pain. On-target and off-target mechanisms mediating TRK inhibitor resistance may occur. Next-generation TRK inhibitors, such as selitrectinib, repotrectinib, and taletrectinib, are available on ongoing clinical trials and address on-target resistance. This review will focus on NTRK fusions and TRK-directed targeted therapy specifically in the context of lung cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号