Crizotinib

克唑替尼
  • 文章类型: Journal Article
    随着下一代测序(NGS)在实体瘤中的广泛应用,间充质到上皮转化因子(MET)重排/融合已在多种癌症类型中得到证实。MET扩增和MET外显子14跳跃突变诱导蛋白质自身磷酸化;然而,MET融合的致病机制和药物敏感性尚不清楚。以下报告描述了被诊断患有带有TFG-MET基因融合体的鳞状肺癌的患者的临床病例。体外试验表明,由于TFG-MET重排,MET磷酸化和致癌能力,两者均被克唑替尼治疗抑制.患者接受克唑替尼治疗,导致持续超过17个月的部分缓解。总的来说,细胞分析和我们的病例报告强调了MET融合作为实体瘤个性化靶向治疗的预测性生物标志物的潜力。
    With the widespread use of next-generation sequencing (NGS) for solid tumors, mesenchymal-to-epithelial transition factor (MET) rearrangement/fusion has been confirmed in multiple cancer types. MET amplification and MET exon 14 skipping mutations induce protein autophosphorylation; however, the pathogenic mechanism and drug sensitivity of MET fusion remain unclear. The following report describes the clinical case of a patient diagnosed with squamous lung cancer bearing a TFG-MET gene fusion. In vitro assays demonstrated MET phosphorylation and oncogenic capacity due to the TFG-MET rearrangement, both of which were inhibited by crizotinib treatment. The patient was treated with crizotinib, which resulted in sustained partial remission for more than 17 months. Collectively, cellular analyses and our case report emphasize the potential of MET fusion as a predictive biomarker for personalized target therapy for 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
    间变性甲状腺癌(ATC)是最致命的人类癌症之一,占甲状腺癌的<2%。ATC的治疗靶点以间变性淋巴瘤激酶(ALK)重排为代表,参与肿瘤生长。克唑替尼是ALK的口服小分子酪氨酸激酶抑制剂,MET,和ROS1激酶,ALK阳性非小细胞肺癌。直到现在,文献中尚未报道克唑替尼对“原代人ATC细胞”(pATCs)与转化纹状体蛋白(STRN)-ALK融合的作用。在这项研究中,我们的目的是在体外使用STRN-ALK获得pATC,并评估克唑替尼的体外抗肿瘤作用.甲状腺手术样本来自12名ATC患者和6名对照(接受了甲状旁腺切除术)。总共获得了10/12pATC培养物,其中2与转化的STRN-ALK融合(17%)。克唑替尼抑制增殖,迁移,3/10pATC培养物中的侵袭和凋亡增加(其中2个带有/1个无STRN-ALK),特别是那些有STRN-ALK的。此外,克唑替尼显着抑制AF细胞(从原代ATC细胞获得的连续细胞系)的增殖。总之,在体外临床前研究中,克唑替尼的抗肿瘤活性已在人pATCs(与STRN-ALK)中显示,为这些患者未来的临床评估开辟了道路。
    Anaplastic thyroid cancer (ATC) is one of the deadliest human cancers and represents <2% of thyroid carcinomas. A therapeutic target for ATC is represented by anaplastic lymphoma kinase (ALK) rearrangements, involved in tumor growth. Crizotinib is an oral small-molecule tyrosine kinase inhibitor of the ALK, MET, and ROS1 kinases, approved in ALK-positive non-small cell lung cancer. Until now, the effect of crizotinib in \"primary human ATC cells\" (pATCs) with transforming striatin (STRN)-ALK fusion has not been reported in the literature. In this study, we aimed to obtain pATCs with STRN-ALK in vitro and evaluate the in vitro antineoplastic action of crizotinib. Thyroid surgical samples were obtained from 12 ATC patients and 6 controls (who had undergone parathyroidectomy). A total of 10/12 pATC cultures were obtained, 2 of which with transforming STRN-ALK fusion (17%). Crizotinib inhibited proliferation, migration, and invasion and increased apoptosis in 3/10 pATC cultures (2 of which with/1 without STRN-ALK), particularly in those with STRN-ALK. Moreover, crizotinib significantly inhibited the proliferation of AF cells (a continuous cell line obtained from primary ATC cells). In conclusion, the antineoplastic activity of crizotinib has been shown in human pATCs (with STRN-ALK) in preclinical studies in vitro, opening the way to future clinical evaluation in these patients.
    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
    阐明CYP3A4活性抑制和基因多态性对克唑替尼代谢的影响。建立了克唑替尼的酶孵育系统,和Sprague-Dawley大鼠用于体内实验。使用LC-MS/MS定量分析物。在筛选122种药物和天然化合物后,原花青素作为克唑替尼代谢抑制剂,表现出93.7%的相对抑制率。大鼠肝微粒体的IC50值为24.53±0.32μM,人肝微粒体的IC50值为18.24±0.12μM。体内研究表明,原花色素显着影响克唑替尼的药代动力学参数。共同给药导致AUC(0-t)显着降低,PF-06260182(克唑替尼的主要代谢产物)的Cmax,和尿代谢比。这种相互作用归因于原花青素对肝微粒体活性的混合型抑制。CYP3A4是克唑替尼的主要代谢酶,其遗传多态性显著影响克唑替尼的药代动力学。动力学数据显示,与野生型CYP3A4.1相比,克唑替尼在26个CYP3A4变体中的相对代谢率范围为13.14%(CYP3A4.12,13)至188.57%(CYP3A4.33)。此外,与野生型相比,原花青素的抑制作用在CYP3A4.12和CYP3A4.33之间有所不同。我们的发现表明原花青素联合给药和CYP3A4基因多态性可以显着影响克唑替尼的代谢。
    To elucidate the impact of CYP3A4 activity inhibition and genetic polymorphism on the metabolism of crizotinib. Enzymatic incubation systems for crizotinib were established, and Sprague-Dawley rats were utilized for in vivo experiments. Analytes were quantified using LC-MS/MS. Upon screening 122 drugs and natural compounds, proanthocyanidins emerged as inhibitor of crizotinib metabolism, exhibiting a relative inhibition rate of 93.7%. The IC50 values were 24.53 ± 0.32 μM in rat liver microsomes and 18.24 ± 0.12 μM in human liver microsomes. In vivo studies revealed that proanthocyanidins markedly affected the pharmacokinetic parameters of crizotinib. Co-administration led to a significant reduction in the AUC(0-t), Cmax of PF-06260182 (the primary metabolite of crizotinib), and the urinary metabolic ratio. This interaction is attributed to the mixed-type inhibition of liver microsome activity by proanthocyanidins. CYP3A4, being the principal metabolic enzyme for crizotinib, has its genetic polymorphisms significantly influencing crizotinib\'s pharmacokinetics. Kinetic data showed that the relative metabolic rates of crizotinib across 26 CYP3A4 variants ranged from 13.14% (CYP3A4.12, 13) to 188.57% (CYP3A4.33) when compared to the wild-type CYP3A4.1. Additionally, the inhibitory effects of proanthocyanidins varied between CYP3A4.12 and CYP3A4.33, when compared to the wild type. Our findings indicate that proanthocyanidins coadministration and CYP3A4 genetic polymorphism can significantly influence crizotinib metabolism.
    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
    小分子激酶抑制剂在现代癌症治疗中起着关键作用。蛋白激酶是癌性肿瘤生长和进展的重要介质,渲染涉及激酶越来越重要的治疗靶点。然而,激酶抑制剂几乎不溶于水,因为它们的疏水芳香性质,通常会降低其可用性和药理功效。与极性基团的直接药物官能化代表了提高药物溶解度的简单策略。可用性,和性能。这里,我们提出了一种策略,使用一步法合成三种示例性激酶抑制药物Ceritinib,用低聚乙二醇(OEG)磷酸酯官能化仲胺,克唑替尼,和Palbociclib。与天然药物相比,这些OEG-前药缀合物在水中表现出优异的溶解度,溶解度增加到190倍。与天然药物相比,缀合物的激酶抑制潜力仅略微降低。我们进一步显示了释放天然药物的OEG-前药的pH依赖性水解。我们观察到在pH3缓慢释放,而缀合物在生理条件(pH7.4)下在96小时内保持稳定。使用共聚焦显微镜,我们验证了药物-OEG缀合物进入HeLa细胞的细胞质的细胞摄取改善,进一步支持我们的通用溶解度方法。
    Small molecular kinase inhibitors play a key role in modern cancer therapy. Protein kinases are essential mediators in the growth and progression of cancerous tumors, rendering involved kinases an increasingly important target for therapy. However, kinase inhibitors are almost insoluble in water because of their hydrophobic aromatic nature, often lowering their availability and pharmacological efficacy. Direct drug functionalization with polar groups represents a simple strategy to improve the drug solubility, availability, and performance. Here, we present a strategy to functionalize secondary amines with oligoethylene glycol (OEG) phosphate using a one-step synthesis in three exemplary kinase inhibiting drugs Ceritinib, Crizotinib, and Palbociclib. These OEG-prodrug conjugates demonstrate superior solubility in water compared to the native drugs, with the solubility increasing up to 190-fold. The kinase inhibition potential is only slightly decreased for the conjugates compared to the native drugs. We further show pH dependent hydrolysis of the OEG-prodrugs which releases the native drug. We observe a slow release at pH 3, while the conjugates remain stable over 96 h under physiological conditions (pH 7.4). Using confocal microscopy, we verify improved cell uptake of the drug-OEG conjugates into the cytoplasm of HeLa cells, further supporting our universal solubility approach.
    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:在第二代ALK酪氨酸激酶抑制剂(ALK-TKIs)的时代,关于进展模式的数据很少,抗性机制,以及随后的ALK阳性(ALK)非小细胞肺癌(NSCLC)的治疗方法。
    方法:回顾性选择本中心的晚期ALK+NSCLC患者。队列1包括接受一线阿来替尼治疗后出现疾病进展的患者(n=20),而队列2包括接受克唑替尼和第二代ALK-TKIs序贯治疗后进展的患者(n=53).寡核苷酸进展定义为在不超过三个病变中发生疾病进展。当患者在放射学进展期间出现新症状或经历先前存在的症状恶化时,确定症状进展。
    结果:与接受克唑替尼治疗的患者相比,第1组的中枢神经系统(CNS)进展和症状性CNS进展的发生率明显较低,比率为15.0%与56.6%(p=0.002)和5.0%与32.1%(p=0.016),分别。共有60.3%(44/73)的患者在第二代ALK-TKI耐药后进行了重复活检和下一代测序,ALK激酶结构域的二次突变成为耐药的主要机制(56.8%)。局部治疗应用于50%的寡进展病例。随后的ALK-TKIs证明无进展生存期(PFS)显着延长(8.6mvs.2.7米,p=0.021,HR=0.43,95CI:0.15-0.85)和长期总生存率(OS)(NA与11.9米,p=0.132,HR=0.50,95CI:0.18-1.25)在具有ALK抗性突变的患者中,与没有这种突变的人相比。对于第二代ALK-TKIs进展后无ALK耐药突变的患者,后续化疗或替代ALK-TKI治疗之间的生存结局无统计学显著差异.
    结论:一线阿来替尼在保护中枢神经系统方面表现出优于克唑替尼的疗效。对于第二代ALK-TKIs耐药后出现ALK耐药突变的患者,应给予适当的敏感ALK-TKI;对于那些没有这种突变的人,化疗或第三代ALK-TKI的选择应基于患者的总体身体健康和个人偏好.
    BACKGROUND: In the era of second-generation ALK tyrosine kinase inhibitors (ALK-TKIs), there was a paucity of data regarding the progression patterns, resistant mechanisms, and subsequent therapeutic approaches for ALK-positive (ALK+) non-small cell lung cancer (NSCLC).
    METHODS: Patients with advanced ALK+ NSCLC were retrospectively selected from our center. Cohort 1 consisted of patients who experienced disease progression after receiving first-line alectinib treatment (n = 20), while Cohort 2 included patients who progressed following sequential treatment with crizotinib and second-generation ALK-TKIs (n = 53). Oligo-progression was defined as the occurrence of disease progression in no more than three lesions. Symptomatic progression was determined when patients developed new symptoms or experienced worsening of pre-existing symptoms during radiological progression.
    RESULTS: The incidence of central nervous system (CNS) progression and symptomatic CNS progression was significantly lower in Cohort 1 compared to patients treated with crizotinib, with rates of 15.0% vs. 56.6% (p = 0.002) and 5.0% vs. 32.1% (p = 0.016), respectively. A total of 60.3% (44/73) patients underwent repeated biopsy and next-generation sequencing subsequent to the second-generation ALK-TKI resistance, with secondary mutation in ALK kinase domain emerging as the predominant mechanism of resistance (56.8%). Local therapy was applied to 50% of oligo-progression cases. Subsequent ALK-TKIs demonstrated significantly prolonged progression-free survival (PFS) (8.6 m vs. 2.7 m, p = 0.021, HR = 0.43, 95%CI: 0.15-0.85) and long-term overall survival (OS) (NA vs. 11.9 m, p = 0.132, HR = 0.50, 95%CI: 0.18-1.25) in patients harboring ALK resistance mutations, compared to those without such mutations. For patients without ALK-resistant mutations following progression on second-generation ALK-TKIs, there was no statistically significant difference in survival outcomes between subsequent chemotherapy or alternative ALK-TKI treatments.
    CONCLUSIONS: First-line alectinib demonstrated superior efficacy in protecting the CNS compared to crizotinib. For patients with ALK-resistant mutations following the resistance to second-generation ALK-TKIs, appropriate sensitive ALK-TKI should be administered; for those without such mutations, the selection of chemotherapy or third-generation ALK-TKI should be based on the patient\'s overall physical health and personal preferences.
    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
    我们介绍了一名70岁从不吸烟的女性患者,患有表皮生长因子受体(EGFR)p.L858R突变的转移性非小细胞肺癌(NSCLC)。厄洛替尼一线治疗三个月后,进展,铂/培美曲塞开始,随后是两年多的回应。进步之后,椎体转移的分子检测显示ROS原癌基因1(ROS1)易位和人表皮生长因子受体2(HER2)p.S310F突变,除了已知的EGFRp.L858R突变。然后克唑替尼导致了17个月的持久反应。从复发性胸腔积液中获得的肿瘤细胞的分子再测试显示不存在ROS1易位,而EGFR和HER2突变仍然存在.阿法替尼被添加到克唑替尼中,联合治疗导致了另一个超过两年的持久反应。患者在最初诊断为转移性NSCLC后超过7年死亡。该病例表明,对转移性NSCLC的重复分子检测可能会发现新的可成药的基因组改变,从而影响患者的管理并改善患者的预后。
    We present the case of a 70-year-old never-smoking female patient with epidermal growth factor receptor (EGFR) p.L858R-mutated metastatic non-small cell lung cancer (NSCLC). After three months of first-line treatment with erlotinib, progression occurred and platinum/pemetrexed was initiated, followed by a response for more than two years. After the progression, the molecular testing of a vertebral metastasis revealed a ROS proto-oncogene 1 (ROS1) translocation and a human epidermal growth factor receptor 2 (HER2) p.S310F mutation, in addition to the known EGFR p.L858R mutation. Crizotinib then led to a durable response of 17 months. The molecular retesting of the tumour cells obtained from the recurrent pleural effusion revealed the absence of the ROS1 translocation, whereas the EGFR and HER2 mutations were still present. Afatinib was added to the crizotinib, and the combination treatment resulted in another durable response of more than two years. The patient died more than 7 years after the initial diagnosis of metastatic NSCLC. This case demonstrates that the repeated molecular testing of metastatic NSCLC may identify new druggable genomic alterations that can impact the patient management and improve the patient outcome.
    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:间变性淋巴瘤激酶(ALK)基因的改变在间变性大细胞淋巴瘤(ALCL)的发病机制中起关键作用。克唑替尼是ALK的小分子竞争性抑制剂,ROS1和MET激酶被批准用于ALK阳性复发或难治性儿科患者,系统性ALCL,ALK阳性不能切除,经常性,或难治性炎性肌纤维母细胞瘤(IMT)。
    方法:来自复发性或难治性实体瘤患儿的Crizotinib数据,IMT,或ALCL纳入分析.所有患者均以每天两次(BID)100至365mg/m2的剂量口服克唑替尼。进行PopPK分析以表征儿科患者的克唑替尼处置。进行暴露-反应(ER)安全性和抗肿瘤分析以表征克唑替尼剂量或暴露与安全性和感兴趣的抗肿瘤活性终点之间的关系。
    结果:群体药代动力学(popPK),ER安全,ER抗肿瘤分析包括98、110和36名儿科患者,分别。具有异速尺度的单室药代动力学模型,一阶消除,具有滞后时间的一阶吸收充分描述了数据。自然对数转换模型预测的克唑替尼AUCss(浓度-时间曲线下的稳态面积)显示出显著的,与≥3级中性粒细胞减少和任何级别视觉障碍呈正相关。克唑替尼剂量与客观缓解率呈正相关。
    结论:在不同年龄或不同类型的肿瘤中,PK没有显著差异。建议基于体表面积(BSA)的剂量根据患者体型差异进行适当调整,以在幼儿和青少年儿科患者中实现相似的全身克唑替尼暴露.除≥3级中性粒细胞外,无骨髓抑制事件与克唑替尼剂量或暴露量有显著关系,提示对于ALK突变的儿科癌症患者,克唑替尼是一种可耐受的治疗方案,与传统化疗方案相比,其血液学毒性较小.所呈现的分析结果支持产品标签中的儿科给药建议。
    BACKGROUND: Alterations in the ALK (anaplastic lymphoma kinase) gene play a critical role in pathogenesis of anaplastic large cell lymphoma (ALCL). Crizotinib is a small molecule competitive inhibitor of ALK, ROS1, and MET kinases and was approved for pediatric patients with ALK-positive relapsed or refractory, systemic ALCL, and ALK-positive unresectable, recurrent, or refractory inflammatory myofibroblastic tumors (IMT).
    METHODS: Crizotinib data from pediatric patients with relapsed or refractory solid tumors, IMT, or ALCL were included in the analyses. All patients received crizotinib orally at doses ranging from 100 to 365 mg/m2 twice daily (BID). PopPK analyses were conducted to characterize crizotinib disposition in pediatric patients. Exposure-response (ER) safety and antitumor analyses were conducted to characterize relationships between crizotinib dose or exposure with safety and antitumor activity endpoints of interest.
    RESULTS: The population pharmacokinetic (popPK), ER safety, and ER antitumor analysis included 98, 110, and 36 pediatric patients, respectively. A one-compartment pharmacokinetic model with allometric scaling, first-order elimination, and first-order absorption with lag time adequately described the data. Natural log-transformed model-predicted crizotinib AUCss (steady-state area under the concentration-time curve) demonstrated a significant, positive relationship with Grade ≥3 NEUTROPENIA and Any Grade VISION DISORDER. Crizotinib dose demonstrated a positive relationship with objective response rate.
    CONCLUSIONS: No significant differences in PK were identified across a wide range of ages or across tumor types, suggesting body surface area (BSA)-based dosing adequately adjusted for differences in patient size to achieve similar systemic crizotinib exposures across young children and adolescent pediatric patients. None of the myelosuppressive events except Grade ≥3 NEUTROPENIA had significant relationships identified with crizotinib dose or exposure, suggesting crizotinib is a tolerable treatment with less hematological toxicity than traditional chemotherapy regimens for pediatric patients with ALK-mutated cancers. Results from the presented analyses support the pediatric dosing recommendations in the product label.
    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:小儿型弥漫性高级别神经胶质瘤(pHGG)是儿童中最常见的恶性脑肿瘤,可以细分为多个实体。激活MET受体酪氨酸激酶的融合基因通常发生在婴儿型半球神经胶质瘤(IHG)中,也发生在其他pHGG中,并与破坏性的发病率和死亡率有关。
    方法:为了确定新的治疗方案,我们建立并表征了两种具有不同MET融合的新型原位小鼠模型。其中包括免疫能力,小鼠同种异体移植模型和患者来源的原位异种移植物(PDOX),来自MET融合IHG患者,该患者的常规治疗和卡博替尼靶向治疗失败。有了这些模型,我们分析了三种MET抑制剂的药效和药代动力学特性,卡马替尼,克唑替尼和卡博替尼,单独或联合放疗。
    结果:卡马替尼在两种模型中都显示出比卡博替尼或克唑替尼更好的脑药代动力学特性和更大的体外和体内功效。PDOX模型概括了患者经历的卡博替尼的不良疗效。相比之下,在两个互补小鼠模型中,卡马替尼与放疗联合治疗可延长生存期并诱导长期无进展生存期.卡马替尼治疗增加了辐射诱导的DNA双链断裂并延迟了其修复。
    结论:我们全面研究了MET抑制和放疗的组合作为MET驱动的pHGG的新治疗选择。我们开创性的临床前数据包包括药代动力学表征,临床结果的概述,多重补充体内研究的结果一致,以及对增加疗效的分子机制的见解。一起来看,我们证明了卡马替尼和放疗的突破性疗效,作为未来临床试验的一个非常有前景的概念.
    BACKGROUND: Pediatric-type diffuse high-grade glioma (pHGG) is the most frequent malignant brain tumor in children and can be subclassified into multiple entities. Fusion genes activating the MET receptor tyrosine kinase often occur in infant-type hemispheric glioma (IHG) but also in other pHGG and are associated with devastating morbidity and mortality.
    METHODS: To identify new treatment options, we established and characterized two novel orthotopic mouse models harboring distinct MET fusions. These included an immunocompetent, murine allograft model and patient-derived orthotopic xenografts (PDOX) from a MET-fusion IHG patient who failed conventional therapy and targeted therapy with cabozantinib. With these models, we analyzed the efficacy and pharmacokinetic properties of three MET inhibitors, capmatinib, crizotinib and cabozantinib, alone or combined with radiotherapy.
    RESULTS: Capmatinib showed superior brain pharmacokinetic properties and greater in vitro and in vivo efficacy than cabozantinib or crizotinib in both models. The PDOX models recapitulated the poor efficacy of cabozantinib experienced by the patient. In contrast, capmatinib extended survival and induced long-term progression-free survival when combined with radiotherapy in two complementary mouse models. Capmatinib treatment increased radiation-induced DNA double-strand breaks and delayed their repair.
    CONCLUSIONS: We comprehensively investigated the combination of MET inhibition and radiotherapy as a novel treatment option for MET-driven pHGG. Our seminal preclinical data package includes pharmacokinetic characterization, recapitulation of clinical outcomes, coinciding results from multiple complementing in vivo studies, and insights into molecular mechanism underlying increased efficacy. Taken together, we demonstrate the groundbreaking efficacy of capmatinib and radiation as a highly promising concept for future clinical trials.
    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
    MET外显子14(METex14)跳跃突变是在大约3-4%的非小细胞肺癌(NSCLC)中观察到的致癌驱动因素。已经报道了导致METex14的几种不同的遗传改变,但罕见突变的临床意义以及MET抑制剂(METi)患者的预后尚未明确。
    本报告介绍了一例转移性NSCLC患者,该患者具有罕见的MET突变景观,尤其包括一种新型METex14突变(R1022L)。在克唑替尼下观察到明显但短暂的疗效,由于早期发生了获得性上和脱靶抗性机制,如METD1246H突变和野生型KRAS扩增。
    我们的案例提供了有关MET罕见致癌变异体及其对METi敏感性的其他数据。酪氨酸激酶抑制剂后肿瘤样品的系统评估对于鉴定可能代表耐药患者的治疗可靶向驱动因素的靶和脱靶机制仍然至关重要。
    UNASSIGNED: MET exon 14 (METex14) skipping mutations are oncogenic drivers observed in approximately 3-4% of non-small cell lung cancers (NSCLC). Several distinct genetic alterations leading to METex14 have been reported but clinical significances of rare mutations are not well defined as well as outcomes of patients upon MET inhibitors (METi).
    UNASSIGNED: This report presents the case of a patient with metastatic NSCLC harboring an uncommon MET mutational landscape including notably a novel METex14 mutation (R1022L). Dramatic but transient efficacy was observed under crizotinib, due to early occurrence of acquired both on- and off-target mechanisms of resistance such as MET D1246H mutation and wild-type KRAS amplification.
    UNASSIGNED: Our case provides additional data on MET rare oncogenic variants and their sensitivity to METi. Systematic assessment of post-tyrosine kinase inhibitor tumor sample remains critical to identify on- and off-target mechanisms that may represent therapeutically targetable drivers in resistant patients.
    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
    与缺乏驱动基因突变的NSCLC病例相比,程序性细胞死亡配体1(PD-L1)更容易在ROS原癌基因1(ROS1)重排的非小细胞肺癌(NSCLC)中表达。先前的研究已经建立了PD-L1表达与EGFR或ALK抑制剂在EGFR或ALK阳性NSCLC中的有效性降低之间的联系。尽管如此,初始PD-L1水平与克唑替尼一线治疗ROS1重排NSCLC的临床影响之间的关系仍不确定.
    从2016年1月至2021年12月,共收集了246例ROS1阳性肿瘤患者。在这些中,82例晚期ROS1重排非小细胞肺癌患者,他们接受克唑替尼作为初始治疗,被选中进行研究。该研究主要旨在评估客观缓解率(ORR)和无进展生存期(PFS)。其次评估疾病控制率(DCR)和总生存期(OS)。
    在82例晚期ROS1重排非小细胞肺癌患者中,38例显示PD-L1阳性,细分为11个高表达水平和27个低表达水平,其余44例没有PD-L1表达。所有纳入患者的ORR为80.5%。在不同PD-L1表达状态的ROS1重排的NSCLC患者中,未观察到ORR的统计学差异。然而,PD-L1阴性组(100%)和高表达组(90.9%)的DCR差异有统计学意义(p=0.04).PD-L1阴性组的中位PFS为26.4个月,低表达组的16.6,和13.7高表达组(p=0.001)。此外,PD-L1阴性组和阳性组的中位PFS也存在显著的统计学差异(p=0.02).对于整个研究人群来说,中位OS为53.0个月(95%CI43.8-62.2).在PD-L1阴性组中,中位OS达到57.2个月,与PD-L1阳性组的53.0个月相比,差异缺乏统计学意义(p=0.43)。
    我们的结果表明,对于接受克唑替尼作为一线治疗的ROS1阳性NSCLC患者,PD-L1表达可能是PFS而不是OS的阴性预后标志物。
    UNASSIGNED: Programmed cell death ligand 1 (PD-L1) is more readily expressed in ROS proto-oncogene 1 (ROS1) rearranged non-small cell lung cancer (NSCLC) compared to NSCLC cases lacking driver gene mutations. Prior research has established a link between PD-L1 expression and reduced effectiveness of EGFR or ALK inhibitors in EGFR or ALK-positive NSCLC. Nonetheless, the relationship between initial PD-L1 levels and the clinical impact of first-line crizotinib therapy in ROS1-rearranged NSCLC is still uncertain.
    UNASSIGNED: From January 2016 to December 2021, a total of 246 patients with ROS1 positive tumors were collected. Out of these, 82 patients with advanced ROS1-rearranged NSCLC, who were treated with crizotinib as their initial therapy, were selected for the study. The study aimed primarily to evaluate the objective response rate (ORR) and progression-free survival (PFS), and secondarily to assess disease control rate (DCR) and overall survival (OS).
    UNASSIGNED: Of the 82 advanced ROS1-rearranged NSCLC patients, 38 exhibited PD-L1 positivity, subdivided into 11 with high and 27 with low expression levels, while the remaining 44 showed no PD-L1 expression. The ORR for all included patients was 80.5%. No statistically significant variance in ORR was observed among ROS1-rearranged NSCLC patients across differing PD-L1 expression statuses. However, there was a statistically significant difference in DCR between PD-L1 negative group (100%) and high expression group (90.9%) (p=0.04). The median PFS spanned 26.4 months for the PD-L1 negative group, 16.6 for the low expression group, and 13.7 for the high expression group (p=0.001). Additionally, a notable statistical disparity was also observed in median PFS between the PD-L1 negative and positive groups (p=0.02). For the entire study population, the median OS was 53.0 months (95% CI 43.8 - 62.2). In the PD-L1-negative group, the median OS reached 57.2 months, compared to 53.0 months in the PD-L1-positive group, a difference lacking statistical significance (p=0.43).
    UNASSIGNED: Our results suggest that for ROS1-positive NSCLC patients receiving crizotinib as first-line therapy, PD-L1 expression may serve as a negative prognostic marker for PFS rather than OS.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号