tyrosine kinase

酪氨酸激酶
  • 文章类型: Journal Article
    造血细胞激酶(Hck),属于Src激酶家族的非受体酪氨酸激酶,与许多人类疾病的发病机理有着错综复杂的联系,与癌症有特别明显的关联。Hck不仅直接影响扩散,迁移,和癌细胞的凋亡,但也与JAK/STAT相互作用,MEK/ERK,PI3K/AKT,CXCL12/CXCR4等途径。Hck还影响肿瘤微环境以促进癌症的发作和进展。本文探讨了Hck在多种实体瘤中的功能作用和调控机制。此外,它探讨了Hck在血液恶性肿瘤中的意义。这篇综述总结了Hck抑制剂的研究现状,其中大多数处于研究的临床前阶段。值得注意的是,这些抑制剂主要用于白血病的治疗管理,它们的组合潜力为未来的研究指明了有希望的途径。总之,这篇综述强调了Hck在实体瘤中的作用机制的意义。这种见解对于理解有关Hck的当前研究趋势至关重要:针对Hck的靶向治疗在恶性肿瘤的诊断和治疗中显示出巨大的前景。进一步研究Hck在癌症中的作用,再加上特异性抑制剂的开发,有可能彻底改变癌症治疗方法。
    Hematopoietic cell kinase (Hck), a non-receptor tyrosine kinase belonging to the Src kinase family, is intricately linked to the pathogenesis of numerous human diseases, with a particularly pronounced association with cancer. Hck not only directly impacts the proliferation, migration, and apoptosis of cancer cells but also interacts with JAK/STAT, MEK/ERK, PI3K/AKT, CXCL12/CXCR4, and other pathways. Hck also influences the tumor microenvironment to facilitate the onset and progression of cancer. This paper delves into the functional role and regulatory mechanisms of Hck in various solid tumors. Additionally, it explores the implications of Hck in hematological malignancies. The review culminates with a summary of the current research status of Hck inhibitors, the majority of which are in the pre-clinical phase of investigation. Notably, these inhibitors are predominantly utilized in the therapeutic management of leukemia, with their combinatorial potential indicating promising avenues for future research. In conclusion, this review underscores the significance of the mechanism of Hck in solid tumors. This insight is crucial for comprehending the current research trends regarding Hck: targeted therapy against Hck shows great promise in both diagnosis and treatment of malignant tumors. Further investigation into the role of Hck in cancer, coupled with the development of specific inhibitors, has the potential to revolutionize approaches to cancer treatment.
    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
    酪氨酸酶(TYR)抑制剂是非常重要的,因为它们抑制酪氨酸酶活性,它的抑制作用对皮肤护理至关重要,抗癌药物,以及水果和蔬菜的抗褐变。这项工作提出了一种使用氨氯地平制备新的合成酪氨酸酶抑制剂的新的经济途径(4)。设计了新型偶联物6(a-o),合成,并以光谱分析为特征,包括傅里叶变换红外和低和高分辨率质谱。纯化的化合物4与各种醛和酮5(a-o)在甲醇中在60℃-90℃回流5-8小时。这项研究以逐步的方式修饰了该药物,以开发作为酪氨酸酶抑制剂的治疗特性。基于光谱和分析数据建立合成的配体6(a-o)的结构。对合成的化合物6(a-o)进行酪氨酸酶的筛选。曲酸作为标准。所制备的化合物6(a-o)对酪氨酸酶具有良好的抑制潜力。化合物6o,6b,6f,和6k描绘了优异的抗酪氨酸酶活性。化合物6k,IC50值为5.34±0.58µM,与标准曲酸(IC506.04±0.11µM)一样有效,在所有合成化合物6(a-o)中脱颖而出。通过PatchDock评估缀合物6(a-o)的计算机模拟研究。化合物6k显示8,999的结合亲和力得分和-219.66kcal/mol的原子接触能(ACE)值。结构-活性关系表明,二氢吡啶核和一些活化基团在亚苄基亚胺部分的邻位和对位的存在是酪氨酸酶活性良好的主要因素。化合物6k可作为药物修饰的先导化合物作为酪氨酸酶抑制剂用于皮肤护理,抗癌药物,水果和蔬菜的抗褐变。
    Tyrosinase (TYR) inhibitors are very significant as they inhibit enzyme tyrosinase activity, and its inhibition is vital for skin care, anticancer medication, and antibrowning of fruits and vegetables. This work presents a novel and economical route for the preparation of new synthetic tyrosinase inhibitors using amlodipine (4). The novel conjugates 6 (a-o) were designed, synthesized, and characterized by spectroscopic analyses, including Fourier transform infrared and low- and high-resolution mass spectroscopy. The purified compound 4 was refluxed with various aldehydes and ketones 5 (a-o) for 5-8 h in methanol at 60°C-90°C. This research modified the drug in a step-by-step manner to develop therapeutic properties as a tyrosinase inhibitor. The structures of synthesized ligands 6 (a-o) were established based on spectral and analytical data. The synthesized compounds 6 (a-o) were screened against tyrosinase enzyme. Kojic acid was taken as standard. All the prepared compounds 6 (a-o) have good inhibition potential against the enzyme tyrosinase. Compounds 6o, 6b, 6f, and 6k depicted excellent antityrosinase activity. Compound 6k, with an IC50 value of 5.34 ± 0.58 µM, is as potent as the standard kojic acid (IC50 6.04 ± 0.11 µM), standing out among all synthesized compounds 6 (a-o). The in silico studies of the conjugates 6 (a-o) were evaluated via PatchDock. Compound 6k showed a binding affinity score of 8,999 and an atomic contact energy (ACE) value of -219.66 kcal/mol. The structure-activity relationship illustrated that the presence of dihydropyridine nuclei and some activating groups at the ortho and para positions of the benzylideneimine moiety is the main factor for good tyrosinase activity. The compound 6k could be used as a lead compound for drug modification as a tyrosinase inhibitor for skin care, anticancer medication, and antibrowning for fruits and vegetables.
    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
    Janus激酶(JAKs)家族与JAK信号转导和转录信号通路的激活因子相关,在各种细胞过程的调节中起着至关重要的作用。JAK的构象变化是激活的基本步骤,影响多个细胞内信号通路。然而,从非活性激酶到活性激酶的过渡过程仍然是个谜。这项研究旨在研究JAK1的静电特性和过渡状态,以完全激活催化活性酶。为了实现这一目标,对抑制/激活的全长JAK1的结构进行建模,并计算在不同位置具有酪氨酸激酶(TK)结构域的JAK1的能量,和Dijkstra的方法被用来找到能量上最平滑的路径。通过比较激酶失活P733L和S703I突变的能量最平滑路径,提供了对这些突变导致JAK1负调节或正调节的原因的评估。我们的能量分析表明JAK1的激活是热力学自发的,在激活的初始步骤中由能量屏障产生的抑制作用,特别是TK结构域从抑制的四点一的释放,Ezrin,Radixin,Moesin-PK腔。总的来说,这项工作提供了对TK易位的潜在途径和JAK1激活机制的见解。
    The family of Janus Kinases (JAKs) associated with the JAK-signal transducers and activators of transcription signaling pathway plays a vital role in the regulation of various cellular processes. The conformational change of JAKs is the fundamental steps for activation, affecting multiple intracellular signaling pathways. However, the transitional process from inactive to active kinase is still a mystery. This study is aimed at investigating the electrostatic properties and transitional states of JAK1 to a fully activation to a catalytically active enzyme. To achieve this goal, structures of the inhibited/activated full-length JAK1 were modelled and the energies of JAK1 with Tyrosine Kinase (TK) domain at different positions were calculated, and Dijkstra\'s method was applied to find the energetically smoothest path. Through a comparison of the energetically smoothest paths of kinase inactivating P733L and S703I mutations, an evaluation of the reasons why these mutations lead to negative or positive regulation of JAK1 are provided. Our energy analysis suggests that activation of JAK1 is thermodynamically spontaneous, with the inhibition resulting from an energy barrier at the initial steps of activation, specifically the release of the TK domain from the inhibited Four-point-one, Ezrin, Radixin, Moesin-PK cavity. Overall, this work provides insights into the potential pathway for TK translocation and the activation mechanism of JAK1.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Review
    Innate nucleic acid sensing is a ubiquitous and highly conserved immunological process, which is pivotal for monitoring and responding to pathogenic invasion and cellular damage, and central to host defense, autoimmunity, cell fate determination and tumorigenesis. Tyrosine phosphorylation, a major type of post-translational modification, plays a critical regulatory role in innate immune sensing pathway. Core members of nucleic acid sensing signaling pathway, such as cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS), stimulator of interferon genes (STING), and TANK binding kinase 1 (TBK1), are all subject to activity regulation triggered by tyrosine phosphorylation, thereby affecting the host antiviral defense and anti-tumor immunity under physiological or pathological conditions. This review summarizes the recent advances in research on tyrosine kinases and tyrosine phosphorylation in regulation of nucleic acid sensing. The function and potential applications of targeting tyrosine phosphorylation in anti-tumor immunity is disussed to provide insights for understanding and expanding new anti-tumor strategies.
    核酸天然免疫识别是一种普遍存在且高度保守的免疫应答机制,负责监测和响应病原体入侵或组织损伤,进而在宿主防御、自身免疫反应、细胞命运决定以及肿瘤发生发展中发挥关键作用。酪氨酸磷酸化作为一类主要的蛋白质翻译后修饰机制,在核酸识别通路中发挥重要调控功能。其中,介导核酸免疫识别通路的核心成员环鸟苷酸-腺苷酸合成酶(cGAS)、干扰素基因刺激因子(STING)以及TANK结合激酶1(TBK1)等蛋白均受到酪氨酸磷酸化修饰引发的活性调控,进而影响生理或病理条件下宿主的抗病毒防御和抗肿瘤免疫能力。本文综述了酪氨酸激酶和酪氨酸磷酸化修饰在核酸免疫识别中的调控作用及研究现状,讨论了靶向酪氨酸磷酸化在抗肿瘤免疫中的功能和潜在应用,以期为理解并拓展全新的抗肿瘤手段提供思路。.
    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
    胰腺癌(PC)是一种恶性程度最高的肿瘤,生存率低。这主要是由于不可避免的化学抗性。功能失调的酪氨酸激酶(TKs)和长链非编码RNA(lncRNAs)影响PC的耐药性和预后。这里,我们总结了TKs或lncRNAs介导耐药性和其他恶性表型的机制。我们还讨论了lncRNAs发挥致癌或肿瘤抑制作用和不同的机制,包括lncRNA蛋白/microRNAs介导耐药性。此外,我们强调lncRNAs是TKs介导耐药性的上游调节因子。最后,我们展示了TKs(AXL,EGFR,IGF1R,和MET),临床试验,和lncRNAs(LINC00460,PVT1,HIF1A-AS1)。在未来,TKs和lncRNAs可能成为诊断和预后的生物标志物或药物靶标,以克服PC的耐药性。
    Pancreatic cancer (PC) is one of the most malignant tumors with a dismal survival rate, this is primarily due to inevitable chemoresistance. Dysfunctional tyrosine kinases (TKs) and long non-coding RNAs (lncRNAs) affect the drug resistance and prognosis of PC. Here, we summarize the mechanisms by which TKs or lncRNAs mediate drug resistance and other malignant phenotypes. We also discuss that lncRNAs play oncogenic or tumor suppressor roles and different mechanisms including lncRNA-proteins/microRNAs to mediate drug resistance. Furthermore, we highlight that lncRNAs serve as upstream regulators of TKs mediating drug resistance. Finally, we display the clinical significance of TKs (AXL, EGFR, IGF1R, and MET), clinical trials, and lncRNAs (LINC00460, PVT1, HIF1A-AS1). In the future, TKs and lncRNAs may become diagnostic and prognostic biomarkers or drug targets to overcome the drug resistance of PC.
    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
    晚期肝细胞癌(HCC)是一个巨大的公共卫生问题,可治愈的治疗选择有限。阿西替尼,口服酪氨酸激酶抑制剂,是血管内皮生长因子受体(VEGFR)1、2和3的有效和选择性的第二代抑制剂。发现这种抗血管生成药物在各种实体瘤中具有有希望的活性,包括晚期肝癌。目前,然而,没有相关的综述文章总结了阿西替尼在晚期HCC中的确切作用。在这次审查中,24项符合条件的研究(临床试验中的7项研究,八项实验研究,和9项临床试验)被纳入进一步评估。纳入的随机或单臂II期试验表明,与安慰剂治疗晚期HCC相比,阿西替尼不能延长总生存期。但观察到无进展生存期和肿瘤进展时间的改善.实验研究表明,阿西替尼在HCC中的生化作用可能受其相关基因和影响的信号级联(例如VEGFR2/PAK1,CYP1A2,CaMKII/ERK,Akt/mTor,和miR-509-3p/PDGFRA)。FDA批准索拉非尼联合nivolumab(PD-1/PD-L1的抑制剂)作为治疗晚期HCC的一线方案。由于阿西替尼和索拉非尼都是酪氨酸激酶抑制剂以及VEGFR抑制剂,阿西替尼联合抗PDL-1/PD-1抗体也可能在晚期HCC的抗肿瘤作用中表现出巨大潜力。本文综述了阿西替尼在晚期HCC中的临床应用现状和分子机制。通过结合阿西替尼和其他治疗方法在晚期肝癌中走向临床应用,在不久的将来仍需要更多的研究。
    Advanced hepatocellular carcinoma (HCC) is a formidable public health problem with limited curable treatment options. Axitinib, an oral tyrosine kinase inhibitor, is a potent and selective second-generation inhibitor of vascular endothelial growth factor receptor (VEGFR) 1, 2, and 3. This anti-angiogenic drug was found to have promising activity in various solid tumors, including advanced HCC. At present, however, there is no relevant review article that summarizes the exact roles of axitinib in advanced HCC. In this review, 24 eligible studies (seven studies in the ClinicalTrials, eight experimental studies, and nine clinical trials) were included for further evaluation. The included randomized or single-arm phase II trials indicated that axitinib could not prolong the overall survival compared to the placebo for the treatment of advanced HCC, but improvements in progression free survival and time to tumor progression were observed. Experimental studies showed that the biochemical effects of axitinib in HCC might be regulated by its associated genes and affected signaling cascades (e.g. VEGFR2/PAK1, CYP1A2, CaMKII/ERK, Akt/mTor, and miR-509-3p/PDGFRA). FDA approved sorafenib combined with nivolumab (an inhibitor of PD-1/PD-L1) as the first line regimen for the treatment of advanced HCC. Since both axitinib and sorafenib are tyrosine kinase inhibitors as well as the VEGFR inhibitors, axitinib combined with anti-PDL-1/PD-1 antibodies may also exhibit tremendous potential in anti-tumoral effects for advanced HCC. The present review highlights the current clinical applications and the molecular mechanisms of axitinib in advanced HCC. To move toward clinical applications by combining axitinib and other treatments in advanced HCC, more studies are still warranted in the near future.
    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:慢性粒细胞白血病(CML)是一种由骨髓造血干细胞克隆性增殖形成的恶性肿瘤。BCR-ABL融合蛋白,在90%以上的患者中发现,是发现抗CML药物的重要目标。到目前为止,伊马替尼是第一个被FDA批准用于治疗CML的BCR-ABL酪氨酸激酶抑制剂(TKI)。然而,耐药性问题的出现有很多原因,尤其是T135I突变,BCR-ABL的“看门人”。目前,目前临床上尚无长期有效且副作用小的药物。
    方法:本研究拟结合人工智能技术和细胞生长曲线,寻找靶向BCR-ABL的新型TKIs,对T315I突变蛋白具有高抑制活性,细胞毒性,流式细胞术和westernblot实验。
    结果:发现所得化合物能杀死白血病细胞,对BaF3/T315I细胞有较好的抑制作用。4号化合物可以诱导细胞周期停滞,引起自噬和凋亡,抑制BCR-ABL酪氨酸激酶的磷酸化,STAT5和Crkl蛋白。
    结论:结果表明,所筛选的化合物可作为先导化合物进行进一步研究,以发现理想的慢性粒细胞白血病治疗药物。
    BACKGROUND: Chronic myeloid leukemia (CML) is a kind of malignant tumor formed by the clonal proliferation of bone marrow hematopoietic stem cells. BCR-ABL fusion protein, found in more than 90% of patients, is a vital target for discovering anti- CML drugs. Up to date, imatinib is the first BCR-ABL tyrosine kinase inhibitor (TKI) approved by the FDA for treating CML. However, the drug resistance problems appeared for many reasons, especially the T135I mutation, a \"gatekeeper\" of BCR-ABL. Currently, there is no long-term effective and low side effect drug in clinical.
    METHODS: This study intends to find novel TKIs targeting BCR-ABL with high inhibitory activity against T315I mutant protein by combining artificial intelligence technology and cell growth curve, cytotoxicity, flow cytometry and Western blot experiments.
    RESULTS: The obtained compound was found to kill leukemia cells, which had good inhibitory efficacy in BaF3/T315I cells. Compound no 4 could induce cell cycle arrest, cause autophagy and apoptosis, and inhibit the phosphorylation of BCR-ABL tyrosine kinase, STAT5 and Crkl proteins.
    CONCLUSIONS: The results indicated that the screened compound could be used as a lead compound for further research to discover ideal chronic myeloid leukemia therapeutic drugs.
    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
    急性髓细胞性白血病(AML)是一种高度异质性的血液恶性肿瘤。具有FLT3突变的AML患者往往具有高复发率和不良预后,因此FLT3基因已成为AML治疗的重要靶点,并开发了一系列FLT3抑制剂。根据FLT3抑制剂的特点,可分为第一代FLT3抑制剂和第二代FLT3抑制剂。到目前为止,共有8种FLT3抑制剂已经进行了临床试验,只有3种被批准用于治疗AML患者,包括Midostourin,Quizartinib和Gilteritinib。FLT3抑制剂联合标准化疗可提高患者的应答率;在后续维持治疗中,FLT3抑制剂还可以降低疾病复发率,改善患者整体预后。然而,由骨髓微环境引起的主要耐药性,以及由其他突变引起的继发性耐药可能导致FLT3抑制剂的疗效差。对于这样的患者,FLT3抑制剂与其他药物联合应用可减少耐药的发生,提高患者的后续疗效。本文就FLT3抑制剂在AML患者的临床研究及FLT3耐药患者的治疗现状进行综述,为临床医师提供参考。
    Acute myeloid leukemia (AML) is a highly heterogeneous hematological malignancy. AML patients with FLT3 mutations tend to have a high relapse rate and poor outcome, so FLT3 gene has become an important target for AML treatment, and a series of FLT3 inhibitors have been developed. According to the characteristics of FLT3 inhibitors, they can be divided into first-generation FLT3 inhibitors and second-generation FLT3 inhibitors. So far, totally eight FLT3 inhibitors have been undergone clinical trials and only three were approved for the treatment of AML patients, including Midostourin, Quizartinib and Gilteritinib. FLT3 inhibitors can improve the response rate of patients by combining with standard chemotherapy; in the follow-up maintenance treatment, FLT3 inhibitors can also reduce the disease recurrence rate and improve the overall prognosis of patients. However, the primary drug resistance caused by the bone marrow microenvironment, as well as secondary resistance caused by other mutations may result in poor efficacy of FLT3 inhibitors. For such patients, the combination of FLT3 inhibitor with other drugs may reduce the occurrence of drug resistance and improve the subsequent efficacy of patients. This article reviews the current status of FLT3 inhibitors in clinical research of AML patients and the treatment of FLT3-resistant patients to provide reference for clinicians.
    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
    非受体酪氨酸激酶FER的异常过表达已在各种卵巢癌衍生的肿瘤细胞中被报道,并且是患者生存的不良预后因素。它在肿瘤细胞迁移和侵袭中起着至关重要的作用,以激酶依赖性和非依赖性两种方式同时作用,这是不容易被传统的酶抑制剂抑制。然而,蛋白水解靶向嵌合体(PROTACs)技术通过同时靶向酶和支架功能,提供优于传统基于活性的抑制剂的疗效.因此,在这项研究中,我们报告了两种PROTAC化合物的开发,这些化合物以依赖cereblon的方式促进了强大的FER降解。两种PROTAC降解剂的性能都优于FDA批准的药物,布吉替尼,卵巢癌细胞运动抑制。重要的是,这些PROTAC化合物还降解在人类肿瘤样品中鉴定的多种致癌FER融合蛋白。这些结果为应用PROTAC策略拮抗具有FER激酶异常表达的卵巢癌和其他类型癌症的细胞运动性和侵袭性奠定了实验基础,并强调PROTAC是靶向具有多种肿瘤促进功能的蛋白质的优越策略。
    Aberrant overexpression of nonreceptor tyrosine kinase FER (Fps/Fes Related) has been reported in various ovarian carcinoma-derived tumor cells and is a poor prognosis factor for patient survival. It plays an essential role in tumor cell migration and invasion, acting concurrently in both kinase-dependent and -independent manners, which is not easily suppressed by conventional enzymatic inhibitors. Nevertheless, the PROteolysis-TArgeting Chimera (PROTAC) technology offers superior efficacy over traditional activity-based inhibitors by simultaneously targeting enzymatic and scaffold functions. Hence in this study, we report the development of two PROTAC compounds that promote robust FER degradation in a cereblon-dependent manner. Both PROTAC degraders outperform a Food and Drug Administration-approved drug, brigatinib, in ovarian cancer cell motility suppression. Importantly, these PROTAC compounds also degrade multiple oncogenic FER fusion proteins identified in human tumor samples. These results lay an experimental foundation to apply the PROTAC strategy to antagonize cell motility and invasiveness in ovarian and other types of cancers with aberrant expression of FER kinase and highlight PROTACs as a superior strategy for targeting proteins with multiple tumor-promoting functions.
    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
    未经授权:c-MET是各种癌症的重要治疗靶点;然而,中华人民共和国目前仅零售一种特异性c-MET抑制剂。我们的临床前研究揭示了HS-10241抑制c-MET的高选择性。这项第一阶段的研究旨在评估安全性,耐受性,药代动力学,选择性c-MET抑制剂(HS-10241)在晚期实体瘤患者中的抗肿瘤活性。
    UNASSIGNED:局部晚期或转移性实体瘤患者口服单剂量或多剂量HS-10241,每天一次或每天两次,连续21天,其中包括以下六个方案:每天100毫克,200毫克,每天一次,400毫克,每天一次,600毫克,每天一次,200毫克,每天两次,每天两次300毫克。治疗一直持续到疾病进展,不可接受的毒性,或治疗终止。主要终点是剂量限制性毒性和最大耐受剂量(MTD)的发生率。次要终点包括安全性,耐受性,药代动力学,和药效学。
    UNASSIGNED:共有27例晚期非小细胞肺癌患者接受HS-10241,3例患者在每天一次600mgHS-10241治疗后观察到剂量限制性毒性。每日一次给药,MTD为400毫克,每天两次给药,最大安全递增剂量为300毫克,未达到MTD。恶心(48.1%,13of27),疲劳(37.0%,10of27),贫血(33.3%,27个中的9个)是三个最常见的治疗引起的不良事件。每天一次400毫克,Css,最大值为5076ng/mL,稳态曲线下面积为39,998h×ng/mL。MET阳性的患者(n=5)(MET外显子14跳跃,MET扩增,和MET免疫组织化学3+)已确认部分反应(n=1)或疾病稳定(n=3),疾病控制率为80.0%。
    UNASSIGNED:选择性c-MET抑制剂HS-10241在晚期非小细胞肺癌中具有良好的耐受性和临床活性,尤其是MET阳性的患者。此外,本研究阐述了HS-10241在癌症患者中的治疗潜力。
    UNASSIGNED: c-MET is an important therapeutic target for various cancers; however, the People\'s Republic of China currently retails only one specific c-MET inhibitor. Our preclinical study has revealed the high selectivity of HS-10241 to suppress c-MET. This phase 1 study aims to evaluate the safety, tolerability, pharmacokinetics, and antitumor activity of the selective c-MET inhibitor (HS-10241) in patients with advanced solid tumors.
    UNASSIGNED: Patients with locally advanced or metastatic solid tumors orally received a single or multiple dose of HS-10241 once daily or twice daily for 21 consecutive days, which included the following six regimens: 100 mg once daily, 200 mg once daily, 400 mg once daily, 600 mg once daily, 200 mg twice daily, and 300 mg twice daily. The treatment continued until disease progression, unacceptable toxicity, or treatment termination. The primary end point was the incidence of dose-limiting toxicity and maximal tolerated dose (MTD). Secondary end points included safety, tolerability, pharmacokinetics, and pharmacodynamics.
    UNASSIGNED: A total of 27 patients with advanced NSCLC received HS-10241, and dose-limiting toxicity was observed in three patients after 600 mg once-daily HS-10241 treatment. For once-daily dosing, MTD was 400 mg, and for twice-daily dosing, the maximal safe escalated dose was 300 mg, and MTD was not reached. Nausea (48.1%, 13 of 27), fatigue (37.0%, 10 of 27), and anemia (33.3%, 9 of 27) are the three most frequent treatment-emergent adverse events. At 400 mg once daily, Css,max was 5076 ng/mL and steady state area under the curve was 39,998 h × ng/mL. Patients (n = 5) with positive MET (MET exon 14-skipping, MET amplified, and MET immunohistochemistry 3+) had confirmed partial responses (n = 1) or stable disease (n = 3), with a disease control rate of 80.0%.
    UNASSIGNED: The selective c-MET inhibitor HS-10241 was well tolerated and had clinical activity in advanced NSCLC, especially in patients with positive MET. Furthermore, this study expounds on the therapeutic potential of HS-10241 in patients with cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号