fibroblast growth factor receptor

成纤维细胞生长因子受体
  • 文章类型: Journal Article
    DNA适体由于其优异的靶结合特异性和热稳定性而作为生物分子的替代形式受到关注,它们也有望用作受体蛋白的人工激动剂。靶向成纤维细胞生长因子(FGFR)受体的DNA适体激动剂TD0,在伤口愈合和再生医学领域发挥着重要作用,已报道诱导细胞反应以及其天然配体。然而,还指出,长期刺激会有一些不同的反应,表明DNA适体激动剂TD0诱导的细胞内信号不同于天然配体。在本文中,我们全面分析了DNA适配体激动剂TD0靶向FGFR1诱导的细胞内信号,并与天然蛋白配体FGF2进行了比较。发现细胞内信号对于短期刺激是高度相似的。另一方面,受体和下游细胞信号在长时间刺激下表现出不同的激活行为。评估DNA适体激动剂TD0和FGF2在培养基中的稳定性和持续活性表明配体稳定性在适当调节细胞应答中可能是重要的。
    DNA aptamers have attracted attention as an alternative modality for biomolecules due to their excellent target binding specificity and thermal stability, and they are also expected to be applied as artificial agonists for receptor proteins. DNA aptamer agonist TD0 targeting the receptor of fibroblast growth factor (FGFR), which plays an important role in the fields of wound healing and regenerative medicine, has been reported to induce cellular responses as well as its native ligands. However, it was also noted that there were some different responses upon long-term stimulation, suggesting that the intracellular signals induced by DNA aptamer agonist TD0 are different from those of natural ligands. In this paper, we comprehensively analyzed the intracellular signals induced by DNA aptamer agonist TD0 targeting FGFR1, and compared them with those by natural protein ligand FGF2. It was found that the intracellular signals were highly similar for short-term stimulation. On the other hand, the receptor and the downstream cellular signals showed different activation behaviors for long-time stimulation. Evaluating the stability and sustained activity of DNA aptamer agonist TD0 and FGF2 in the medium suggested that ligand stability may be important in properly regulating cellular responses.
    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
    原理:毛囊(HF)和内分泌汗腺(ESG)的重建对于功能性皮肤再生至关重要。在皮肤重建研究中,我们发现包皮来源的表皮细胞单向重建HF类器官,但不是ESG类器官。方法:研究影响ESG和HF命运的关键基因和通路,使用含有ESG胎盘的皮肤和含有HF胎盘的皮肤的转录组分析,通过RT-qPCR和免疫荧光染色对小鼠和大鼠的关键DEGs进行鉴定和验证。随后,通过整合RT-qPCR的一系列方法,重建了成人表皮细胞衍生的类器官,以探测FGF7和FGF10的功能作用和机制,免疫荧光染色,WB,凋亡测定,和通路干扰测定。结果:FGF7亚家族的所有成员都在筛选的关键DEGs中,FGF7和FGF10及其受体FGFR1/FGFR2的差异表达在含ESG胎盘的皮肤和含HF胎盘的皮肤之间得到证实。体内和体外基质胶塞模型显示,FGF7和FGF10均可促进人表皮细胞衍生的类器官向ESG表型类器官的命运转变,FGF7和FGF10具有协同作用,主要通过FGFR1/2-MEK1/2-ERK1/2途径发挥作用。结论:可以操纵成人表皮细胞来重建个性化的HF和ESG,以满足不同的需求。
    Rationale: Reconstruction of hair follicles (HFs) and eccrine sweat glands (ESGs) is essential for functional skin regeneration. In skin reconstruction research, we found that foreskin-derived epidermal cells reconstructed HF organoids unidirectionally, but not ESG organoids. Methods: To investigate key genes and pathways influencing the fate of ESG and HF, a transcriptome profiling of ESG placode-containing skin and HF placode-containing skin was employed, and key DEGs were identified and validated by RT-qPCR and immunofluorescence staining in mice and rats. Subsequently, adult human epidermal cell-derived organoids were reconstructed to probe functional roles and mechanisms of FGF7 and FGF10 by series of approaches integrating RT-qPCR, immunofluorescence-staining, WB, apoptosis assay, and pathway interference assay. Results: All members of FGF7 subfamily were among the key DEGs screened, the differential expression of FGF7 and FGF10 and their receptors FGFR1/FGFR2 was verified between ESG placode-containing skin and HF placode-containing skin. In vivo and in vitro Matrigel plug models showed that both FGF7 and FGF10 promoted fate transition of human epidermal cell-derived organoids to ESG phenotype organoids, FGF7 and FGF10 had a synergistic effect, and mainly function through the FGFR1/2-MEK1/2-ERK1/2 pathway. Conclusions: Adult epidermal cells can be manipulated to reconstruct personalized HF and ESG to meet different needs.
    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
    成纤维细胞生长因子受体(FGFRs)正在成为参与肿瘤发生的关键因素,肿瘤微环境重塑和对靶向治疗的获得性抵抗。Pemigatinib是选择性靶向异常FGFR1、FGFR2和FGFR3的酪氨酸激酶抑制剂。Pemigatinib现已被批准用于晚期胆管癌(CCA),但数据表明其他肿瘤组织型表现出FGFR改变。因此假设其在其他癌症环境中的潜在疗效。本系统综述,根据PRISMA指南,旨在综合和批判性地解释所有可用的临床前和临床证据的结果,关于培米卡替尼用于癌症。2024年4月,在PubMed进行了广泛的搜索,MEDLINE,和Scopus数据库使用关键字“Pemigatinib”。27项研究最终符合所有纳入标准。Pemigatinib临床前和临床研究中出现的有希望的结果为Pemigatinib扩展到多种实体癌环境铺平了道路。
    Fibroblast Growth Factor Receptors (FGFRs) are emerging as key factors involved in tumorigenesis, tumor microenvironment remodeling and acquired resistance to targeted therapies. Pemigatinib is a Tyrosine-Kinase Inhibitor that selectively targets aberrant FGFR1, FGFR2 and FGFR3. Pemigatinib is now approved for advanced-stage cholangiocarcinoma (CCA) but data suggests that other tumor histotypes exhibit FGFR alterations, thus hypothesizing its potential efficacy in other cancer settings. The present systematic review, based on PRISMA guidelines, aims to synthetize and critically interpret the results of all available preclinical and clinical evidence regarding Pemigatinib use in cancer. In April 2024, an extensive search was performed in PubMed, MEDLINE, and Scopus databases using the keyword \"Pemigatinib\". Twenty-seven studies finally met all inclusion criteria. The promising results emerging from Pemigatinib preclinical and clinical studies pave the way for Pemigatinib extension to multiple solid cancer settings.
    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THOR试验(NCT03390504)队列1中,erdafitinib显示,在FGFR改变≥1次治疗后进展的转移性尿路上皮癌(mUC)患者中,与化疗相比,总生存期(OS中位数为12.1个月比7.8个月)显著延长,死亡风险降低36%(风险比0.64,P=0.005)。包括抗PD-(L)1。目前还没有关于日本亚组结果的报道。
    方法:THOR队列1将患者随机分为erdafitinib每日一次或多西他赛/长春氟宁每3周一次。主要终点是OS。次要终点包括无进展生存期(PFS)和客观缓解率(ORR)。没有为该日本亚组分析设置特定的统计功效。
    结果:在266名随机分组的患者中,27(14erdafitinib;13化疗)是日本人。治疗之间和总体人群的基线特征通常相似,除了更多的男性,降低体重,日本患者中上尿路原发性肿瘤较多。与化疗相比,erdafitinib显示OS改善(中位数25.4对12.4个月),PFS(中位数为8.4个月对2.9个月)和ORR(57.1%对15.4%)。两组所有患者均发生任何级别治疗相关不良事件(AE),但erdafitinib组的3/4级AE和导致停药的AE较低。在日本亚组中没有观察到新的安全性信号。
    结论:在日本亚组中,与化疗相比,erdafitinib显示出改善的生存和反应,没有新的安全问题。这些结果支持erdafitinib作为FGFR改变的日本mUC患者的治疗选择,诊断mUC后应考虑早期FGFR检测.
    BACKGROUND: In the THOR trial (NCT03390504) Cohort 1, erdafitinib demonstrated significantly prolonged overall survival (OS) (median 12.1 versus 7.8 months) and reduced risk of death by 36% (hazard ratio 0.64, P = 0.005) compared with chemotherapy in metastatic urothelial carcinoma (mUC) patients with FGFR alterations who progressed after ≥ 1 prior treatments, including anti-PD-(L)1. There have been no reports of the Japanese subgroup results yet.
    METHODS: THOR Cohort 1 randomized patients to erdafitinib once daily or docetaxel/vinflunine once every 3 weeks. Primary endpoint was OS. Secondary endpoints included progression-free survival (PFS) and objective response rate (ORR). No specific statistical power was set for this Japanese subgroup analysis.
    RESULTS: Of 266 patients randomized, 27 (14 erdafitinib; 13 chemotherapy) were Japanese. Baseline characteristics were generally similar between treatments and to the overall population, except for more males, lower body weight, and more upper tract primary tumors among Japanese patients. Compared with chemotherapy, erdafitinib showed improved OS (median 25.4 versus 12.4 months), PFS (median 8.4 versus 2.9 months) and ORR (57.1% versus 15.4%). Any grade treatment-related adverse events (AEs) occurred in all patients from both arms but Grade 3/4 AEs and AEs leading to discontinuation were lower in the erdafitinib arm. No new safety signals were observed in the Japanese subgroup.
    CONCLUSIONS: In the Japanese subgroup, erdafitinib showed improved survival and response compared to chemotherapy, with no new safety concerns. These results support erdafitinib as a treatment option for Japanese mUC patients with FGFR alterations, and early FGFR testing after diagnosis of mUC should be considered.
    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:Pemigatinib是一种口服,强力,选择性成纤维细胞生长因子受体(FGFR)1-3抑制剂。FIGHT-101,由三部分组成,开放标签,人类第一,I/II期研究(NCT02393248),评价了培米加替尼在晚期实体瘤患者中的疗效。在第1部分和第2部分中,培米替尼单药治疗在FGFR改变的肿瘤中具有可控的安全性和抗肿瘤活性。第三部分(培米加替尼联合治疗)结果在此呈现。
    方法:患者连续或间歇服用吉西他滨和顺铂(pemi/gem/cis),每天一次口服9、13.5或20mg培米替尼,多西他赛(pemi/doc),曲妥珠单抗(pemi/tras),pembrolizumab(pemmi/pembro),或retifanlimab(pemi/reti),无论肿瘤是否被证实为FGFR改变。主要终点是安全性和药效学。次要终点是研究者评估的肿瘤客观反应率(ORR)和药代动力学(PK)。
    结果:在65名登记患者中(pemi/gem/cis,n=8;pemi/doc,n=7;pemi/tras,n=6;pemi/pembro,n=26;pemi/reti,n=18),全部停产。治疗引起的不良事件(TEAE)通常与单个药物AE一致。严重和≥3级TEAE发生在0%-85.7%和33.3%-100.0%的患者中。分别。所有培米加替尼组合显示与单一疗法相当的稳态PK。所有培米加替尼组合的药效学效应,除了pemi/gem/cis,与单一疗法一致。用这种组合观察到较少的FGFR2α磷酸化抑制。ORR(95%置信区间)为37.5%[8.5%至75.5%(pemi/gem/cis)],14.3%[0.4%至57.9%(pemi/doc)],0%(pemi/tras),26.9%[11.6%至47.8%(pemi/pembro)],和11.1%[1.4%至34.7%(PEMI/RETI)]。所有组均有肿瘤缩小的情况。FGFR重排和突变的可评估患者的ORR分别为50%和33%,分别。
    结论:Pemigatinib联合治疗无意外毒性。PK和药效学与培米替尼单药治疗基本一致。Pemi/gem/cis(37.5%)和pemmi/pembro(26.9%)的ORR最高;大多数响应者都有FGFR改变。
    BACKGROUND: Pemigatinib is an oral, potent, selective fibroblast growth factor receptor (FGFR) 1-3 inhibitor. FIGHT-101, a three-part, open-label, first-in-human, phase I/II study (NCT02393248), evaluated pemigatinib in patients with advanced solid tumors. In parts 1 and 2, pemigatinib monotherapy had a manageable safety profile and antitumor activity in FGFR-altered tumors. Part 3 (pemigatinib combination therapies) results are presented here.
    METHODS: Patients received 9, 13.5, or 20 mg oral once-daily pemigatinib on continuous or intermittent schedules with gemcitabine and cisplatin (pemi/gem/cis), docetaxel (pemi/doc), trastuzumab (pemi/tras), pembrolizumab (pemi/pembro), or retifanlimab (pemi/reti) irrespective of whether the tumor was confirmed as FGFR altered. Primary endpoints were safety and pharmacodynamics. Secondary endpoints were investigator-assessed tumor objective response rates (ORRs) and pharmacokinetics (PK).
    RESULTS: Of 65 enrolled patients (pemi/gem/cis, n = 8; pemi/doc, n = 7; pemi/tras, n = 6; pemi/pembro, n = 26; pemi/reti, n = 18), all discontinued. Treatment-emergent adverse events (TEAEs) were generally consistent with individual drug AEs. Serious and grade ≥3 TEAEs occurred in 0%-85.7% and 33.3%-100.0% of patients across treatment groups, respectively. All pemigatinib combinations demonstrated steady-state PK comparable to monotherapy. Pharmacodynamic effects in all pemigatinib combinations, except pemi/gem/cis, were consistent with monotherapy. Less inhibition of FGFR2α phosphorylation was observed with this combination. ORRs (95% confidence interval) were 37.5% [8.5% to 75.5% (pemi/gem/cis)], 14.3% [0.4% to 57.9% (pemi/doc)], 0% (pemi/tras), 26.9% [11.6% to 47.8% (pemi/pembro)], and 11.1% [1.4% to 34.7% (pemi/reti)]. All groups had instances of tumor shrinkage. ORRs in assessable patients with FGFR rearrangements and mutations were 50% and 33%, respectively.
    CONCLUSIONS: Pemigatinib combination therapy showed no unexpected toxicities. PK and pharmacodynamics were mostly consistent with pemigatinib monotherapy. Pemi/gem/cis (37.5%) and pemi/pembro (26.9%) had the highest ORR; most responders had FGFR alterations.
    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
    成纤维细胞生长因子(FGFs)是一个多功能的肽生长因子家族,涉及各种生物学功能。包括细胞生长和分化,胚胎发育,血管生成,和新陈代谢。FGF/FGF受体(FGFR)信号异常与多种疾病如癌症的发病机制有关。代谢性疾病,和炎症性疾病。值得注意的是,巨噬细胞极化,涉及不同的功能表型,在组织修复中起着至关重要的作用,稳态维持,和免疫反应。最近的研究表明,FGF/FGFR信号通路与巨噬细胞的极化密切相关,表明它们可能是与功能失调的巨噬细胞相关疾病的治疗操作的潜在靶标。在这篇文章中,我们提供了结构的概述,函数,和FGFs的下游调控途径,以及FGF信号和巨噬细胞极化之间的串扰。此外,我们总结了利用FGF信号调节巨噬细胞极化的潜在应用。
    Fibroblast growth factors (FGFs) are a versatile family of peptide growth factors that are involved in various biological functions, including cell growth and differentiation, embryonic development, angiogenesis, and metabolism. Abnormal FGF/FGF receptor (FGFR) signaling has been implicated in the pathogenesis of multiple diseases such as cancer, metabolic diseases, and inflammatory diseases. It is worth noting that macrophage polarization, which involves distinct functional phenotypes, plays a crucial role in tissue repair, homeostasis maintenance, and immune responses. Recent evidence suggests that FGF/FGFR signaling closely participates in the polarization of macrophages, indicating that they could be potential targets for therapeutic manipulation of diseases associated with dysfunctional macrophages. In this article, we provide an overview of the structure, function, and downstream regulatory pathways of FGFs, as well as crosstalk between FGF signaling and macrophage polarization. Additionally, we summarize the potential application of harnessing FGF signaling to modulate macrophage polarization.
    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
    成纤维细胞生长因子及其受体(FGFR)在人类生长和肿瘤发生中起着重要作用。在成年人中,治疗性FGFR抑制剂已成功对抗携带体细胞FGFR突变的肿瘤.在儿科患者中,测试这些抗肿瘤FGFR抑制剂疗法的试验正在进行中,最近的几份报告表明适度的积极回应。在这里,我们报告了一名患有FGFR1突变神经胶质瘤的青春期前儿童的意外结局,该儿童成功接受了FDA批准的erdafitinib治疗,一种泛FGFR抑制剂被批准用于治疗膀胱肿瘤。在用erdafitinib治疗时,患者经历了快速的骨骼和长骨过度生长,导致脊柱后凸,让人想起先天性功能丧失FGFR3突变的患者。我们利用患者建立的正常真皮成纤维细胞作为替代模型来证明胰岛素样生长因子1(IGF-1),是骨骼和组织发育生长的重要因素,可以激活厄达非替尼处理的细胞中的PI3K/AKT途径,但不能激活MAPK/ERK途径。IGF-I激活的PI3K/AKT信号通过促进细胞存活来拯救正常成纤维细胞免受erdafitinib的细胞毒性作用。我们,因此,假设IGF-I激活的P13K/AKT信号传导可能继续促进生长儿童的骨伸长,但不是成年人,用治疗性泛FGFR抑制剂治疗。重要的是,因为激活的MAPK信号抵消骨伸长,我们进一步推测pan-FGFR抑制剂对MAPK通路的长期阻断,与包括IGF-1在内的生长促进因子的作用一起,可以解释我们的青春期前患者在全身治疗使用pan-FGFR抑制剂期间遭受的异常骨骼和轴向生长.进一步的研究,以找到更有针对性的,和/或适当的剂量,在我们的年轻患者中观察到的泛FGFR抑制剂治疗对于避免意外的脱靶效应至关重要。
    Fibroblast growth factors and their receptors (FGFR) have major roles in both human growth and oncogenesis. In adults, therapeutic FGFR inhibitors have been successful against tumors that carry somatic FGFR mutations. In pediatric patients, trials testing these anti-tumor FGFR inhibitor therapeutics are underway, with several recent reports suggesting modest positive responses. Herein, we report an unforeseen outcome in a pre-pubescent child with an FGFR1-mutated glioma who was successfully treated with FDA-approved erdafitinib, a pan-FGFR inhibitor approved for treatment of Bladder tumors. While on treatment with erdafitinib, the patient experienced rapid skeletal and long bone overgrowth resulting in kyphoscoliosis, reminiscent of patients with congenital loss-of-function FGFR3 mutations. We utilized normal dermal fibroblast cells established from the patient as a surrogate model to demonstrate that insulin-like growth factor 1 (IGF-1), a factor important for developmental growth of bones and tissues, can activate the PI3K/AKT pathway in erdafitinib-treated cells but not the MAPK/ERK pathway. The IGF-I-activated PI3K/AKT signaling rescued normal fibroblasts from the cytotoxic effects of erdafitinib by promoting cell survival. We, therefore, postulate that IGF-I-activated P13K/AKT signaling likely continues to promote bone elongation in the growing child, but not in adults, treated with therapeutic pan-FGFR inhibitors. Importantly, since activated MAPK signaling counters bone elongation, we further postulate that prolonged blockage of the MAPK pathway with pan-FGFR inhibitors, together with actions of growth-promoting factors including IGF-1, could explain the abnormal skeletal and axial growth suffered by our pre-pubertal patient during systemic therapeutic use of pan-FGFR inhibitors. Further studies to find more targeted, and/or appropriate dosing, of pan-FGFR inhibitor therapeutics for children are essential to avoid unexpected off-target effects as was observed in our young patient.
    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:成纤维细胞生长因子受体2(FGFR2)融合和重排是胆管癌(CCA)中临床可行的基因组改变。Pemigatinib是一种选择性的,强力,口服FGFR1-3抑制剂,并在先前治疗的患者中证明了疗效,FIGHT-202中FGFR2改变的晚期/转移性CCA(NCT02924376)。我们报告了延长随访期的最终结果。
    方法:多中心,开放标签,单臂,II期FIGHT-202研究纳入了年龄≥18岁的患者,这些患者先前已治疗过FGFR2融合或重排的晚期/转移性CCA(队列A),其他FGF/FGFR改变(队列B),或无FGF/FGFR改变(队列C)。患者在21天的周期内接受每日一次口服培米加替尼13.5mg(2周,1周休息),直到疾病进展或不可接受的毒性。主要终点是由独立审查委员会根据RECISTv1.1评估的队列A的客观缓解率(ORR);次要终点包括缓解持续时间(DOR),无进展生存期(PFS),总生存期(OS),和安全。
    结果:FIGHT-202招募了147名患者(队列A,108;队列B,20;队列C,17;未经证实的FGF/FGFR改变,2).最后分析,145人(98.6%)因疾病进展而停止治疗(71.4%),患者停药(8.2%),或不良事件(不良事件;6.8%)。中位随访时间为45.4个月。队列A的ORR为37.0%(95%置信区间27.9%至46.9%);在3例和37例患者中观察到完全和部分缓解,分别。中位DOR为9.1(6.0-14.5)个月;中位PFS和OS为7.0(6.1-10.5)个月和17.5(14.4-22.9)个月,分别。最常见的治疗引起的AE(TEAE)是高磷血症(58.5%),脱发(49.7%),腹泻(47.6%)。总的来说,15例(10.2%)患者出现TEAE导致pemigatinib停药;肠梗阻和急性肾损伤(各n=2)发生频率最高。
    结论:在先前接受过治疗的患者中,Pemigatinib表现出持久的缓解和OS延长,AE可控,在FIGHT-202的延长随访期内,晚期/转移性CCA伴FGFR2改变。
    BACKGROUND: Fibroblast growth factor receptor 2 (FGFR2) fusions and rearrangements are clinically actionable genomic alterations in cholangiocarcinoma (CCA). Pemigatinib is a selective, potent, oral inhibitor of FGFR1-3 and demonstrated efficacy in patients with previously treated, advanced/metastatic CCA with FGFR2 alterations in FIGHT-202 (NCT02924376). We report final outcomes from the extended follow-up period.
    METHODS: The multicenter, open-label, single-arm, phase II FIGHT-202 study enrolled patients ≥18 years old with previously treated advanced/metastatic CCA with FGFR2 fusions or rearrangements (cohort A), other FGF/FGFR alterations (cohort B), or no FGF/FGFR alterations (cohort C). Patients received once-daily oral pemigatinib 13.5 mg in 21-day cycles (2 weeks on, 1 week off) until disease progression or unacceptable toxicity. The primary endpoint was objective response rate (ORR) in cohort A assessed as per RECIST v1.1 by an independent review committee; secondary endpoints included duration of response (DOR), progression-free survival (PFS), overall survival (OS), and safety.
    RESULTS: FIGHT-202 enrolled 147 patients (cohort A, 108; cohort B, 20; cohort C, 17; unconfirmed FGF/FGFR alterations, 2). By final analysis, 145 (98.6%) had discontinued treatment due to progressive disease (71.4%), withdrawal by patient (8.2%), or adverse events (AEs; 6.8%). Median follow-up was 45.4 months. The ORR in cohort A was 37.0% (95% confidence interval 27.9% to 46.9%); complete and partial responses were observed in 3 and 37 patients, respectively. Median DOR was 9.1 (6.0-14.5) months; median PFS and OS were 7.0 (6.1-10.5) months and 17.5 (14.4-22.9) months, respectively. The most common treatment-emergent AEs (TEAEs) were hyperphosphatemia (58.5%), alopecia (49.7%), and diarrhea (47.6%). Overall, 15 (10.2%) patients experienced TEAEs leading to pemigatinib discontinuation; intestinal obstruction and acute kidney injury (n = 2 each) occurred most frequently.
    CONCLUSIONS: Pemigatinib demonstrated durable response and prolonged OS with manageable AEs in patients with previously treated, advanced/metastatic CCA with FGFR2 alterations in the extended follow-up period of FIGHT-202.
    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:胆管癌(CCA)是一种侵袭性肿瘤,治疗选择有限,尤其是在二线或后期治疗中。靶向成纤维细胞生长因子受体(FGFR)2最近已成为具有FGFR2融合的CCA患者的有希望的治疗选择。这项研究调查了作为口服有效的FGFR1-3抑制剂的他司格替尼的抗肿瘤活性,在临床前FGFR2驱动的CCA模型中。
    方法:使用表达FGFR2融合蛋白的NIH/3T3细胞作为FGFR2驱动的CCA模型,在体外和体内检查了塔舒替尼的抗肿瘤活性,并在体内使用CCA患者来源的异种移植模型。通过与FGFR1的共晶结构分析,与FGFR2的手动复杂模型分析以及与FGFR2的结合动力学分析,阐明了他格替尼的分子作用机理。此外,在接受FGFR抑制剂治疗的CCA患者中,评估了针对获得性耐药FGFR2突变的细胞抑制活性.
    结果:Tasurgratinib通过在体外和体内抑制FGFR信号通路,在临床前FGFR2驱动的CCA模型中显示出抗肿瘤活性。此外,基于细胞的靶标接合试验表明,他司格替尼对FGFR2突变具有有效的抑制活性,如N549H/K,是CCA中的主要获得性突变。我们还证实,他司格替尼表现出与FGFR2的快速缔合和缓慢解离动力学,与ATP结合位点和邻近区域结合,并采用Asp-Phe-Gly(DFG)-“in”构象。
    结论:这些数据证明了他司格替尼在FGFR2驱动的CCA中的治疗潜力,并提供了分子机制见解,以了解其在接受FGFR抑制剂治疗的CCA患者中对继发性FGFR2耐药突变的独特抑制谱。
    OBJECTIVE: Cholangiocarcinoma (CCA) is an aggressive tumor with limited treatment options especially in 2nd line or later treatments. Targeting fibroblast growth factor receptor (FGFR) 2 has recently emerged as a promising treatment option for patients with CCA harboring FGFR2-fusion. This study investigated the antitumor activities of tasurgratinib as an orally available FGFR1-3 inhibitor, in preclinical FGFR2-driven CCA models.
    METHODS: Antitumor activities of tasurgratinib were examined in vitro and in vivo using NIH/3T3 cells expressing FGFR2-fusion as FGFR2-driven CCA models, and in vivo using a CCA patient-derived xenograft model. The molecular mechanism of action of tasurgratinib was elucidated through co-crystal structure analysis with FGFR1, manual complex model analysis with FGFR2, and binding kinetics analysis with FGFR2. Furthermore, the cell-based inhibitory activities against acquired resistant FGFR2 mutations in patients with CCA treated with FGFR inhibitors were evaluated.
    RESULTS: Tasurgratinib showed antitumor activity in preclinical FGFR2-driven CCA models by inhibiting the FGFR signaling pathway in vitro and in vivo. Furthermore, cell-based target engagement assays indicated that tasurgratinib had potent inhibitory activities against FGFR2 mutations, such as N549H/K, which are the major acquired mutations in CCA. We also confirmed that tasurgratinib exhibited fast association and slow dissociation kinetics with FGFR2, binding to the ATP-binding site and the neighboring region, and adopting an Asp-Phe-Gly (DFG)-\"in\" conformation.
    CONCLUSIONS: These data demonstrate the therapeutic potential of tasurgratinib in FGFR2-driven CCA and provide molecular mechanistic insights into its unique inhibitory profile against secondary FGFR2 resistance mutations in patients with CCA treated with FGFR inhibitors.
    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
    免疫检查点抑制剂(ICIs)对于尿路上皮癌(UC)治疗至关重要。成纤维细胞生长因子受体(FGFR)改变,作为UC中常见的致癌驱动因素,据报道,通过上调FGFR信号驱动UC免疫微环境的T细胞耗尽,这表明FGFR改变可能导致对ICIs的反应降低。此外,选择性pan-FGFR抑制剂在临床试验中显示出更好的临床益处,表明FGFR已通过抑制FGFR信号传导成为关键的治疗靶标。本研究旨在通过1963年UC患者评估FGFR改变的UC患者和FGFR野生型UC患者之间的预后和对ICI的反应,并为UC的个性化精准治疗和联合治疗提供新的见解。
    Immune checkpoint inhibitors (ICIs) are essential for urothelial carcinoma (UC) treatment. Fibroblast growth factor receptor (FGFR) alterations, as common oncogenic drivers in UC, have been reported to drive T cell depletion of UC immune microenvironment via up-regulating FGFR signaling, which indicated FGFR alterations potentially result in reduced response to ICIs. In addition, the selective pan-FGFR inhibitor showed better clinical benefit in clinical trials, indicating FGFR has emerged as critical therapeutic target via inhibiting FGFR signaling. The present study aims to evaluate prognosis and response to ICIs between FGFR-altered UC patients and FGFR-wildtype UC patients via 1963 UC patients and offers new insights into personalized precision therapy and combination therapy for UC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号