Tyrosine kinase inhibitor

酪氨酸激酶抑制剂
  • 文章类型: Journal Article
    化疗耐药是癌症治疗的主要障碍,通常导致疾病进展和不良结局。它通过各种机制产生,如基因突变,药物外排泵,增强DNA修复,和肿瘤微环境的变化。这些过程使癌细胞尽管化疗也能存活,强调需要新的策略来克服耐药性和提高治疗效果。克唑替尼,第一代多靶点激酶抑制剂,FDA批准用于ALK阳性或ROS1阳性非小细胞肺癌(NSCLC)的治疗,难治性炎性(ALK)阳性肌纤维母细胞瘤(IMT)和复发性/难治性ALK阳性间变性大细胞淋巴瘤(ALCL)。克唑替尼以两种对映体形式存在:(R)-克唑替尼及其镜像,(S)-克唑替尼。假定R-异构体负责进行此处审查的各种方法S-异构体,另一方面,显示出对DNA修复机制重要的酶MTH1的强烈抑制作用。研究表明,克唑替尼是一种有效的多激酶抑制剂,靶向多种激酶,如c-Met,native/T315IBcr/Abl,JAK2其作用机制涉及ATP结合的竞争性抑制和变构抑制,特别是在Bcr/Abl。克唑替尼与聚ADP核糖聚合酶抑制剂(PARP)联合使用时显示出协同作用,尤其是在携带BRCA基因突变的卵巢癌中。此外,crizotinib靶向许多p53突变癌症的一个关键的脆弱性。与它的野生型对应物不同,p53突变体促进癌细胞存活.克唑替尼可导致p53突变体的降解,使这些癌细胞对DNA损伤物质敏感并引发细胞凋亡。有趣的是,其他报道表明,克唑替尼具有抗菌活性,靶向革兰氏阳性菌。此外,它对耐药菌株有活性。总之,克唑替尼通过多种机制发挥抗肿瘤作用,包括抑制激酶和恢复药物敏感性。强调了克唑替尼在联合治疗中的潜力,特别是在p53突变高患病率的癌症中,如三阴性乳腺癌(TNBC)和高级别浆液性卵巢癌(HGSOC)。
    Chemoresistance is a major obstacle in cancer treatment, often leading to disease progression and poor outcomes. It arises through various mechanisms such as genetic mutations, drug efflux pumps, enhanced DNA repair, and changes in the tumor microenvironment. These processes allow cancer cells to survive despite chemotherapy, underscoring the need for new strategies to overcome resistance and improve treatment efficacy. Crizotinib, a first-generation multi-target kinase inhibitor, is approved by the FDA for the treatment of ALK-positive or ROS1-positive non-small cell lung cancer (NSCLC), refractory inflammatory (ALK)-positive myofibroblastic tumors (IMTs) and relapsed/refractory ALK-positive anaplastic large cell lymphoma (ALCL). Crizotinib exists in two enantiomeric forms: (R)-crizotinib and its mirror image, (S)-crizotinib. It is assumed that the R-isomer is responsible for the carrying out various processes reviewed here The S-isomer, on the other hand, shows a strong inhibition of MTH1, an enzyme important for DNA repair mechanisms. Studies have shown that crizotinib is an effective multi-kinase inhibitor targeting various kinases such as c-Met, native/T315I Bcr/Abl, and JAK2. Its mechanism of action involves the competitive inhibition of ATP binding and allosteric inhibition, particularly at Bcr/Abl. Crizotinib showed synergistic effects when combined with the poly ADP ribose polymerase inhibitor (PARP), especially in ovarian cancer harboring BRCA gene mutations. In addition, crizotinib targets a critical vulnerability in many p53-mutated cancers. Unlike its wild-type counterpart, the p53 mutant promotes cancer cell survival. Crizotinib can cause the degradation of the p53 mutant, sensitizing these cancer cells to DNA-damaging substances and triggering apoptosis. Interestingly, other reports demonstrated that crizotinib exhibits anti-bacterial activity, targeting Gram-positive bacteria. Also, it is active against drug-resistant strains. In summary, crizotinib exerts anti-tumor effects through several mechanisms, including the inhibition of kinases and the restoration of drug sensitivity. The potential of crizotinib in combination therapies is emphasized, particularly in cancers with a high prevalence of the p53 mutant, such as triple-negative breast cancer (TNBC) and high-grade serous ovarian cancer (HGSOC).
    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:酪氨酸激酶抑制剂(TKIs)索拉非尼和乐伐替尼代表了肝移植(LT)后肝细胞癌(HCC)复发患者的首选一线全身治疗。索拉非尼和乐伐替尼,多年来,HCC患者在临床研究中显示出越来越多的总体生存率。相比之下,TKIs下LT术后HCC复发患者的总生存期数据很少,仅限于小型回顾性研究.在这次回顾中,多中心研究,我们研究了TKI治疗的疗效和免疫抑制对肝癌患者肝移植后复发的影响。
    方法:回顾性数据来自德国和奥地利的四个移植中心。我们纳入了2007年至2020年间接受TKI治疗的LT术后HCC复发患者。
    结果:总计,我们分析了46例肝移植后HCC复发患者的数据。最常见的潜在肝病是丙型肝炎,占52.2%。中位复发时间为11.8个月(0-117.7个月)。21例患者(45.7%)肝脏移植受累,36例(78.3%)在初次诊断复发时发生肝外转移,与肺是最常见的影响(n=25,54.3%)。在总数中,54.3%(n=25)的患者最初接受了局部治疗;39例(85.8%)和7例(15.2%)患者接受了索拉非尼和乐伐替尼,分别,作为一线全身治疗。整个队列的中位总生存期为10.9个月(95%置信区间(95%CI)6.9-14.9个月),中位无进展生存期为5.7个月(95%CI2.0-9.4个月)。
    结论:由于肝移植史被认为是免疫治疗的禁忌症,肝移植后HCC复发患者的预后仍然很差。
    BACKGROUND: The tyrosine kinase inhibitors (TKIs) sorafenib and lenvatinib represent the first-line systemic therapy of choice for patients with hepatocellular carcinoma (HCC) recurrence after liver transplantation (LT). Under sorafenib and lenvatinib, HCC patients have shown increasingly improved overall survival in clinical studies over the years. In contrast, data on overall survival for patients with HCC recurrence after LT under TKIs are scarce and limited to small retrospective series. In this retrospective, multicenter study, we investigated the efficacy of TKI therapy and the influence of immunosuppression in patients with HCC recurrence after LT.
    METHODS: Retrospective data were collected from four transplant centers from Germany and Austria. We included patients with HCC recurrence after LT between 2007 and 2020 who were treated with a TKI.
    RESULTS: In total, we analyzed data from 46 patients with HCC recurrence after LT. The most common underlying liver disease was hepatitis C, accounting for 52.2%. The median time to relapse was 11.8 months (range 0-117.7 months). The liver graft was affected in 21 patients (45.7%), and 36 patients (78.3%) had extrahepatic metastases at initial diagnosis of recurrence, with the lung being the most commonly affected (n = 25, 54.3%). Of the total, 54.3% (n = 25) of the patients were initially treated locally; 39 (85.8%) and 7 (15.2%) patients received sorafenib and lenvatinib, respectively, as first-line systemic therapy. Median overall survival of the whole cohort was 10.9 months (95% confidence interval (95% CI) 6.9-14.9 months) and median progression free survival was 5.7 months (95% CI 2.0-9.4 months) from treatment initiation.
    CONCLUSIONS: Since history of liver transplantation is considered a contraindication for immunotherapy, prognosis of patients with HCC recurrence after LT remains poor.
    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:Lenvatinib是一种酪氨酸激酶抑制剂,可以改善甲状腺癌和肝细胞癌患者的无进展生存期。然而,它受到不良心血管事件的限制,包括高血压和心功能不全.内质网应激的激活参与心肌细胞凋亡。
    目的:本研究旨在通过靶向激活转录因子6(ATF6)来确认来伐替尼的心脏毒性是否与内质网应激有关,需要肌醇酶1α(IRE1α)和蛋白激酶RNA样ER激酶(PERK)信号通路。
    方法:雄性C57/BL6小鼠灌胃给予30mg/kg/天的乐伐替尼。心电图(ECG)和超声心动图用于检测心律失常和心功能。新生大鼠心肌细胞用乐伐替尼处理48h。细胞计数试剂盒(CCK8),2',7'-二氯二氢荧光素二乙酸酯(H2DCFHDA),Hoechst33258和二氢罗丹明123分别用于评估细胞活力,活性氧(ROS)的水平,核形态改变和线粒体膜电位(MMP)水平。
    结果:Lenvatinib在舒张期和收缩期显着降低了左心室的后壁厚度,但对左心室射血分数(LVEF,%).此外,lenvatinib大大延长了校正的QT间期(QTc)并改变了心肌细胞的形态。在小鼠心脏切片中没有发现纤维化的显著差异。Lenvatinib上调凋亡相关蛋白表达。此外,乐伐替尼增加了ERS相关蛋白的表达(GRP78,CHOP,和ATF6)和增强的PERK磷酸化。在新生大鼠心肌细胞中,乐伐替尼显著降低心肌细胞活力并诱导细胞凋亡。此外,ROS产量增加,MMP减少。类似于老鼠的实验,lenvatinib导致凋亡相关蛋白和ERS相关蛋白上调,并增加PERK和IRE1α的磷酸化水平。
    结论:Lenvatinib诱导的心脏毒性与ERS通过靶向ATF6,IRE1α诱导的细胞凋亡有关,和PERK信号通路。
    BACKGROUND: Lenvatinib is a tyrosine kinase inhibitor that can improve progression-free survival in patients with thyroid cancer and hepatocellular carcinoma. However, it is limited by adverse cardiovascular events, including hypertension and cardiac dysfunction. Activation of endoplasmic reticulum stress is involved in cardiomyocyte apoptosis.
    OBJECTIVE: This study aimed to confirm whether the cardiotoxicity of lenvatinib is associated with endoplasmic reticulum stress by targeting the activating transcription factor 6 (ATF6), inositol- requiring enzyme 1α (IRE1α) and protein kinase RNA-like ER kinase (PERK) signaling pathways.
    METHODS: Male C57/BL6 mice were intragastric administration with 30 mg/kg/day lenvatinib. Electrocardiography (ECG) and echocardiography were used to detect arrhythmias and cardiac function. Neonatal rat cardiomyocytes were treated with lenvatinib for 48h. Cell counting kit (CCK8), 2´,7´-dichlorodihydrofluoresceine diacetate (H2DCFHDA), Hoechst 33258 and dihydrorhodamine 123 were respectively used for evaluating cell viability, the level of reactive oxygen species (ROS), nuclear morphological changes and mitochondrial membrane potential (MMP) level.
    RESULTS: Lenvatinib remarkably decreased the posterior wall thickness of left ventricle during diastole and systole but caused little decrease to the left ventricular ejection fraction (LVEF, %). Furthermore, lenvatinib greatly prolonged the corrected QT interval (QTc) and altered the morphology of cardiomyocytes. No dramatic difference in fibrosis was found in mouse cardiac slices. Lenvatinib upregulates apoptosis-related protein expression. In addition, lenvatinib increased ERS-related protein expression (GRP78, CHOP, and ATF6) and enhanced PERK phosphorylation. In neonatal rat cardiac myocytes, lenvatinib markedly decreased the viability of cardiomyocytes and induced apoptosis. Furthermore, ROS production increased and MMP decreased. Similar to the mice experiment, lenvatinib caused upregulation of apoptosis-related and ERS-related proteins and increased the phosphorylation levels of PERK and IRE1α.
    CONCLUSIONS: Lenvatinib-induced cardiotoxicity is associated with ERS-induced apoptosis by targeting the ATF6, IRE1α, and PERK signaling pathways.
    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
    系统性肥大细胞增多症(SM)是一种罕见的骨髓增殖性肿瘤,其特征是克隆性肥大细胞异常增殖和浸润不同组织。肥大细胞的不受控制的增殖和活化引发血管活性和炎症介质的释放,导致一系列的全身症状.大约95%的SM来自KIT基因的功能获得突变,特别是在816密码子,这突出了它在SM中的重要作用,使其成为一个有吸引力的治疗靶点。尽管KIT阴性SM非常罕见,文献中记录的病例数量的增加使其成为这种疾病的一个有趣的方面。报告的KIT阴性SM的临床表现变化很大,但许多类似于KIT阳性SM。针对KIT的治疗选择已经改变了KIT阳性SM的游戏规则,然而,它们在KIT阴性SM中的作用仍然存在争议。本报告旨在通过介绍两例KIT阴性SM来进一步了解KIT阴性SM,其中之一是对KIT靶向治疗有反应,并对现有文献中报道的病例进行分析。
    Systemic mastocytosis (SM) is a rare type of myeloproliferative neoplasm characterized by abnormal proliferation and infiltration of different tissue by clonal mast cells. The uncontrolled proliferation and activation of mast cells trigger the release of vasoactive and inflammatory mediators, resulting in a cascade of systemic symptoms. Around 95% of SM arise from a gain-of-function mutation at the KIT gene, specifically at codon 816, which highlights its essential role in SM and makes it an attractive target for therapy. Although KIT-negative SM is exceptionally rare, the increased number of cases documented in the literature makes it an intriguing dimension of this disorder. The reported clinical manifestations of KIT-negative SM are widely variable, but many are similar to KIT-positive SM. KIT-targeted therapeutic options have been a game-changer in KIT-positive SM, however their role in KIT-negative SM remains controversial. This report aimed to further understand KIT-negative SM by presenting two cases of KIT-negative SM, one of which was responsive to KIT-targeted therapy, and analyzing reported cases in the existing literature.
    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:大多数胃肠道间质瘤(GIST)存在c-KIT或PDGFRA突变。酪氨酸激酶抑制剂(TKIs)的施用显著改善了患有GIST的患者的存活率。我们旨在评估台湾晚期或复发性GIST患者的临床结局。
    方法:纳入2010年至2020年确诊的患者。收集的数据包括基线特征,治疗模式,治疗结果,遗传畸变和生存状态。分析无进展生存期(PFS)和总生存期(OS)并用Kaplan-Meier法绘制。Cox回归分析影响生存的预后因素。
    结果:共纳入224例接受TKIs治疗的晚期或复发性GIST患者。所有患者均接受伊马替尼治疗。93例和42例患者接受舒尼替尼和瑞戈非尼治疗,分别。使用伊马替尼治疗的患者48个月PFS和OS分别为50.5%和79.5%,分别。在多变量Cox回归分析中,c-KIT外显子9和PDGFRA突变是PFS不良的预后因素,PDGFRA突变是伊马替尼治疗患者OS不良的预后因素。接受舒尼替尼治疗的患者的中位PFS为12.76个月(95%置信区间(CI),11.01-14.52)。具有c-KIT外显子9突变的患者比具有其他遗传畸变的患者具有更长的PFS。接受regorafenib治疗的患者的中位PFS为7.14个月(95%CI,3.39-10.89)。
    结论:我们展示了接受TKIs治疗的晚期GIST患者的真实临床结果,并确定了突变状态作为患者生存的独立预后因素。
    BACKGROUND: Most gastrointestinal stromal tumors (GISTs) harbor c-KIT or PDGFRA mutations. Administration of tyrosine kinase inhibitors (TKIs) has significantly improved the survival of patients with GISTs. We aimed to evaluate the clinical outcome of advanced or recurrent GIST patients in Taiwan.
    METHODS: Patients diagnosed between 2010 and 2020 were enrolled. The collected data included baseline characteristics, treatment pattern, treatment outcome, genetic aberrations and survival status. Progression-free survival (PFS) and overall survival (OS) were analyzed and plotted with the Kaplan-Meier method. Cox regression analysis was used to analyze the prognostic factors of survival.
    RESULTS: A total of 224 patients with advanced or recurrent GISTs treated with TKIs were enrolled. All patients received imatinib treatment. Ninety-three and 42 patients received sunitinib and regorafenib treatment, respectively. The 48-month PFS and OS rates for patients treated with imatinib were 50.5% and 79.5%, respectively. c-KIT exon 9 and PDGFRA mutations were prognostic factors for a poor PFS and PDGFRA mutation was a prognostic factor for a poor OS in patients treated with imatinib in multivariate Cox regression analysis. The median PFS of patients who received sunitinib treatment was 12.76 months (95% confidence interval (CI), 11.01-14.52). Patients with c-KIT exon 9 mutations had a longer PFS than those with other genetic aberrations. The median PFS of patients treated with regorafenib was 7.14 months (95% CI, 3.39-10.89).
    CONCLUSIONS: We present real-world clinical outcomes for advanced GIST patients treated with TKIs and identify mutational status as an independent prognostic factor for patient survival.
    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:在肿瘤治疗中,蛋白酪氨酸激酶抑制剂(TKIs)已被广泛使用。然而,TKI的疗效明显受到耐药性的影响。因此,找到克服TKI耐药性的有效解决方案变得至关重要。活性氧(ROS)是一组高活性分子,在包括TKI靶向治疗在内的靶向癌症治疗中起重要作用。在这次审查中,我们专注于肿瘤中TKI致死率的ROS相关机制以及调节ROS逆转肿瘤中TKI耐药的策略。
    方法:在癌症的TKI治疗过程中,经常表现出ROS水平升高,可能导致细胞器损伤和细胞死亡,这对TKIs成功根除癌细胞至关重要。然而,值得注意的是,癌细胞可能会启动抵抗途径来保护自己免受ROS诱导的损害,导致TKI抵抗。应对这一挑战涉及阻断这些抗性途径,例如,NRF2-KEAP1轴和保护性自噬,促进ROS在细胞中的积累,从而使耐药癌细胞对TKIs重新敏感。诱导耐药细胞内ROS产生并提供外源性ROS刺激的其他有效方法。
    结论:ROS在TKI根除肿瘤细胞中起关键作用。利用ROS的积累来克服TKI抗性是一种有效且广泛适用的方法。
    BACKGROUND: In tumor treatment, protein tyrosine kinase inhibitors (TKIs) have been extensively utilized. However, the efficacy of TKI is significantly compromised by drug resistance. Consequently, finding an effective solution to overcome TKI resistance becomes crucial. Reactive oxygen species (ROS) are a group of highly active molecules that play important roles in targeted cancer therapy including TKI targeted therapy. In this review, we concentrate on the ROS-associated mechanisms of TKI lethality in tumors and strategies for regulating ROS to reverse TKI resistance in cancer.
    METHODS: Elevated ROS levels often manifest during TKI therapy in cancers, potentially causing organelle damage and cell death, which are critical to the success of TKIs in eradicating cancer cells. However, it is noteworthy that cancer cells might initiate resistance pathways to shield themselves from ROS-induced damage, leading to TKI resistance. Addressing this challenge involves blocking these resistance pathways, for instance, the NRF2-KEAP1 axis and protective autophagy, to promote ROS accumulation in cells, thereby resensitizing drug-resistant cancer cells to TKIs. Additional effective approaches inducing ROS generation within drug-resistant cells and providing exogenous ROS stimulation.
    CONCLUSIONS: ROS play pivotal roles in the eradication of tumor cells by TKI. Harnessing the accumulation of ROS to overcome TKI resistance is an effective and widely applicable approach.
    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
    转化生长因子-β(TGF-β)信号通路失调调节肿瘤干细胞(CSCs)和药物敏感性,而反馈调节机制是如何被劫持到燃料耐药CSC上的,目前还不清楚.通过利用干细胞样耐药特性作为读数的全基因组CRISPR激活筛选,TGF-β受体相关结合蛋白1(TGFBRAP1)被鉴定为TGF-β诱导的正反馈调节因子,其控制对酪氨酸激酶抑制剂(TKIs)的敏感性并促进肝癌干性.通过与TGF-β受体1型(TGFBR1)相互作用并稳定,TGFBRAP1在增强TGF-β信号传导中起重要作用。机械上,TGFβ1与E3泛素连接酶Smurf1/2竞争结合TGFβR1,导致受损的受体聚泛素化和蛋白酶体降解。此外,过度活跃的TGF-β信号传导上调耐药CSC样细胞中TGF-β1的表达,从而构成先前未表征的反馈机制以放大TGF-β信号传导。因此,TGFβ1表达与肝细胞癌(HCC)组织中TGFβR1水平和TGF-β信号活性相关,以及多个HCC队列中的总体生存率和疾病复发。治疗学上,通过选择性抑制剂阻断TGFBR1介导的TGFBR1稳定通过减少CSC减轻Regorafenib耐药性。总的来说,TGF-β信号通路的靶向反馈机制可能是减轻耐药和肝癌干性的可行方法。
    Dysregulation of the transforming growth factor-β (TGF-β) signaling pathway regulates cancer stem cells (CSCs) and drug sensitivity, whereas it remains largely unknown how feedback regulatory mechanisms are hijacked to fuel drug-resistant CSCs. Through a genome-wide CRISPR activation screen utilizing stem-like drug-resistant properties as a readout, the TGF-β receptor-associated binding protein 1 (TGFBRAP1) is identified as a TGF-β-inducible positive feedback regulator that governs sensitivity to tyrosine kinase inhibitors (TKIs) and promotes liver cancer stemness. By interacting with and stabilizing the TGF-β receptor type 1 (TGFBR1), TGFBRAP1 plays an important role in potentiating TGF-β signaling. Mechanistically, TGFBRAP1 competes with E3 ubiquitin ligases Smurf1/2 for binding to TGFΒR1, leading to impaired receptor poly-ubiquitination and proteasomal degradation. Moreover, hyperactive TGF-β signaling in turn up-regulates TGFBRAP1 expression in drug-resistant CSC-like cells, thereby constituting a previously uncharacterized feedback mechanism to amplify TGF-β signaling. As such, TGFBRAP1 expression is correlated with TGFΒR1 levels and TGF-β signaling activity in hepatocellular carcinoma (HCC) tissues, as well as overall survival and disease recurrence in multiple HCC cohorts. Therapeutically, blocking TGFBRAP1-mediated stabilization of TGFBR1 by selective inhibitors alleviates Regorafenib resistance via reducing CSCs. Collectively, targeting feedback machinery of TGF-β signaling pathway may be an actionable approach to mitigate drug resistance and liver cancer stemness.
    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
    铁凋亡是一种依赖于铁的细胞死亡,其特征在于脂质过氧化的发生和活性氧的积累。Ferroptosis已被证明对肝细胞癌(HCC)的治疗进展和耐药性有显著影响,从而突出了其作为可行的治疗靶标的潜力。与正常肝组织相比,在HCC组织中观察到铁凋亡。已发现抑制铁凋亡可增加HCC细胞的活力并降低其对各种抗癌疗法的敏感性。包括化疗,放射治疗,和免疫检查点封锁。直接调节铁凋亡调节剂或诱导脂质活性氧物质过量产生的药物的施用已证明了增强耐药HCC细胞对治疗的反应性的潜力。然而,这种现象背后的确切机制仍然模棱两可。这篇综述全面概述了铁凋亡在提高肝细胞癌(HCC)治疗疗效中的关键作用。这项研究的主要目的是研究利用铁性凋亡作为治疗方法的可行性,以提高HCC治疗的疗效并克服耐药性。
    Ferroptosis is a type of cell death that relies on iron and is distinguished by the occurrence of lipid peroxidation and the buildup of reactive oxygen species. Ferroptosis has been demonstrated to have a significant impact on the advancement and resistance to treatment of hepatocellular carcinoma (HCC), thereby highlighting its potential as a viable therapeutic target. Ferroptosis was observed in HCC tissues in contrast to normal liver tissue. The inhibition of ferroptosis has been found to increase the viability of HCC cells and decrease their susceptibility to various anticancer therapies, including chemotherapy, radiotherapy, and immune checkpoint blockade. The administration of drugs that directly modulate ferroptosis regulators or induce excessive production of lipid-reactive oxygen species has demonstrated the potential to enhance the responsiveness of drug-resistant HCC cells to treatment. However, the precise mechanism underlying this phenomenon remains ambiguous. This review presents a comprehensive overview of the crucial role played by ferroptosis in enhancing the efficacy of treatment for hepatocellular carcinoma (HCC). The main aim of this study is to examine the feasibility of utilizing ferroptosis as a therapeutic approach to improve the efficacy of HCC treatment and overcome drug resistance.
    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
    最近,靶向治疗方法的发展,如基于酪氨酸激酶抑制剂(TKI)的靶向治疗方法极大地改善了癌基因成瘾的晚期非小细胞肺癌(NSCLC)患者的临床结局.同样,放射治疗技术的改进允许向有限数量的转移性靶病变(少持续或少进展)提供高辐射剂量,有限的高剂量正常组织暴露导致低严重毒性率。这篇叙述性综述的目的是概述目前建立的寡转移和寡进展疾病的定义,定义一线和后续一线靶向治疗,以及在这些设置中巩固非侵入性局部消融治疗(LAT)的作用。局部治疗(LT)如放疗(RT)或手术的潜在益处可能表现为转换到随后的全身治疗的整体减少,从而降低了进一步全身传播的风险。进一步的随机临床试验将阐明LT的作用及其与全身靶向治疗相关的正确时机。
    Recently, the development of targeted therapy approaches such as those based on tyrosine kinase inhibitor (TKI) greatly improved the clinical outcomes of patients affected by oncogene addicted advanced non-small cell lung cancer (NSCLC). Similarly, the improvement of radiation therapy techniques has permitted to deliver high radiation doses to a limited number of metastatic target lesions (oligopersistent or oligoprogressive), with limited high-dose normal tissue exposure that leads to low severe toxicity rates. The aim of this narrative review was to provide an overview of the currently established definition of oligometastatic and oligoprogressive disease, to define first line and subsequent lines targeted therapies and the role of consolidative non-invasive local ablative treatments (LATs) in these settings. The potential benefit of local treatment (LT) such as radiotherapy (RT) or surgery might be represented by an overall reduction of switching to subsequent systemic treatments lowering the risk of further systemic dissemination. Further randomized clinical trials will clarify the role of LT and their correct timing in relation to systemic targeted therapies.
    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
    本文基于我们先前的研究,该文件在2023年ASCO年会上发表,并作为会议摘要发表在《临床肿瘤学杂志》上(JCO。2023年;41:e16148。doi:10.1200/JCO.2020.23.41.16_suppl。e16148).抗程序性死亡1/配体-1(PD-1/L1)抗体+抗血管内皮生长因子(VEGF)抗体(A+A)和抗PD-1/L1抗体+VEGF受体(VEGFR)靶向酪氨酸激酶抑制剂(A+T)是不可切除的肝细胞癌的有效一线疗法。然而,缺乏对这两种治疗方法进行正面对比的证据.我们对它们的有效性和安全性进行了网络荟萃分析。
    经过严格的文献研究,确定了6个III期试验进行最终分析,包括IMbrave150、ORIENT-32、COSMIC-312、CARES-310、LEAP-002和REFLECT。实验分为三组:A+A,A+T,和中间参考组。主要终点是总生存期(OS),次要终点包括无进展生存期(PFS),客观反应率(ORR),和治疗相关不良事件(TRAEs)的发生率。OS和PFS的风险比(HR)和95%置信区间(CI),ORR的比值比(OR),并计算所有等级和≥3级TRAE的相对风险(RR)。在贝叶斯框架下,荟萃分析使用索拉非尼作为中间参考.
    等级概率为96%,A+A显示死亡风险降低最大,与A+T无显著差异(HR:0.82,95%CI:0.65-1.04)。A+T在延长PFS和改善ORR方面效果最大,等级概率为77%,但与A+A无统计学差异,就所有TRAE等级(RR:0.91,95%CI:0.82-1.00)而言,A+A比A+T更安全,尤其是那些等级≥3级(RR:0.65,95%CI:0.54-0.77).
    A+A最有可能提供最长的操作系统,而A+T与较高的PFS获益相关,但安全性较低。
    UNASSIGNED: This article is based on our previous research, which was presented at the 2023 ASCO Annual Meeting I and published in Journal of Clinical Oncology as Conference Abstract (JCO. 2023;41:e16148. doi: 10.1200/JCO.2023.41.16_suppl.e16148). Both anti-programmed death 1/ligand-1 (PD-1/L1) antibody + anti-vascular endothelial growth factor (VEGF) antibody (A + A) and anti-PD-1/L1 antibody + VEGF receptor (VEGFR)-targeted tyrosine kinase inhibitor (A + T) are effective first-line therapies for unresectable hepatocellular carcinoma. However, there lacks evidence from head-to-head comparisons between these two treatments. We conducted a network meta-analysis on the efficacy and safety of them.
    UNASSIGNED: After a rigorous literature research, 6 phase III trials were identified for the final analysis, including IMbrave150, ORIENT-32, COSMIC-312, CARES-310, LEAP-002, and REFLECT. The experiments were classified into three groups: A + A, A + T, and intermediate reference group. The primary endpoint was overall survival (OS), and secondary endpoints included progression-free survival (PFS), objective response rate (ORR), and incidence of treatment-related adverse events (TRAEs). Hazard ratio (HR) with 95% confidence intervals (CI) for OS and PFS, odds ratio (OR) for ORR, and relative risk (RR) for all grade and grade ≥3 TRAEs were calculated. Under Bayesian framework, the meta-analysis was conducted using sorafenib as intermediate reference.
    UNASSIGNED: With the rank probability of 96%, A + A showed the greatest reduction in the risk of death, without significant difference from A + T (HR: 0.82, 95% CI: 0.65-1.04). A + T showed the greatest effect in prolonging PFS and improving ORR with the rank probability of 77%, but there were no statistical differences with A + A. A + A was safer than A + T in terms of all grade of TRAEs (RR: 0.91, 95% CI: 0.82-1.00) and particularly in those grade ≥3 (RR: 0.65, 95% CI: 0.54-0.77).
    UNASSIGNED: A + A had the greatest probability of delivering the longest OS, while A + T was correlated with larger PFS benefits at the cost of a lower safety rate.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号