FGFR

FGFR
  • 文章类型: Journal Article
    这项研究调查了FGFR抑制剂futibatinib(FTB)和MEK抑制剂binimetinib(BTB)用于KRASmtNSCLC治疗的组合。开发并验证了一种用于测量大鼠血浆中FTB和BTB浓度的分析方法,遵守USFDA指南。对45μL血浆样品进行液-液萃取,达到6.5-min的运行时间。线性校准曲线范围为2至100ng/mL。日内和日间准确度介于92.06%和100.08%之间。高浓度样品后的四次空白注射解决了明显的残留。不同浓度的萃取回收率平均为92.06%~102.37%。在空白血浆中未检测到显著的内源性干扰。两种药物的LLOQ均为2.0ng/mL。选择性,矩阵效应,稳定性,和稀释完整性满足验收标准。该方法评估了5mg/kg联合治疗中的FTB和BTB相互作用潜力。这些发现为未来的临床试验提供了必要的药代动力学见解。
    This study investigates the combination of FGFR inhibitor futibatinib (FTB) and MEK inhibitor binimetinib (BTB) for KRASmt NSCLC therapy. An analytical method was developed and validated for measuring FTB and BTB concentrations in rat plasma, adhering to USFDA guidelines. Using liquid-liquid extraction on 45-μL plasma samples, a 6.5-min run time was achieved. The linear calibration curve ranged from 2 to 100 ng/mL. Intra-day and inter-day accuracy ranged between 92.06% and 100.08%. Four blank injections post high-concentration samples resolved significant carryover. Extraction recoveries averaged 92.06% to 102.37% across concentrations. No significant endogenous interference was detected in blank plasma. The LLOQ for both drugs was 2.0 ng/mL. Selectivity, matrix effects, stability, and dilution integrity met the acceptance criteria. The method assessed FTB and BTB interaction potential in combination therapy at 5 mg/kg. The findings provide essential pharmacokinetics insights for future clinical trials.
    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
    虽然临床前研究一直暗示FGFR信号传导在乳腺癌(BC)进展中,临床证据不能支持这些发现.FGFR的临床意义可能应该在基质的背景下进行分析,激活或抑制其功能。本综述旨在通过总结有关选定的癌症相关成纤维细胞(CAFs)相关因子的预后和/或预测价值的现有数据来提供这样的背景。这可能直接或间接影响FGFR信令。PubMed(https://pubmed.ncbi.nlm.nih.gov/)和Medline(https://www.nlm.nih.gov/medline/medline_home.html)数据库中搜索了与以下因素的预后和/或预测意义相关的相关文献:CAFs表型标记(αSMA,S100A4/FSP‑1,PDGFR,PDPN和FAP),CAFs衍生的同源FGFR配体(FGF2,FGF5和FGF17)或CAFs旁分泌活性的诱导剂(TGF-β1,HDGF,PDGF,CXCL8,CCL5,CCL2,IL‑6,HH和EGF)均在肿瘤中表达并在血液中循环。总共选择了68篇文章并进行了全面分析。这些发现一致地确定了αSMA的上调,S100A4/FSP‑1,PDGFR,PDPN,HDGF,PDGF,CXCL8,CCL5,CCL2,IL-6,HH和EGF作为BC的不良预后标志物,而对其余标志物的预后价值的评估在研究之间有所不同。数据证实了CAFs特异性特征与BC预后的关联,提示可能需要对基质进行定量和定性分析以评估真实FGFR的临床价值.
    While preclinical studies consistently implicate FGFR‑signalling in breast cancer (BC) progression, clinical evidence fails to support these findings. It may be that the clinical significance of FGFR ought to be analysed in the context of the stroma, activating or repressing its function. The present review aimed to provide such a context by summarizing the existing data on the prognostic and/or predictive value of selected cancer‑associated fibroblasts (CAFs)‑related factors, that either directly or indirectly may affect FGFR‑signalling. PubMed (https://pubmed.ncbi.nlm.nih.gov/) and Medline (https://www.nlm.nih.gov/medline/medline_home.html) databases were searched for the relevant literature related to the prognostic and/or predictive significance of: CAFs phenotypic markers (αSMA, S100A4/FSP‑1, PDGFR, PDPN and FAP), CAFs‑derived cognate FGFR ligands (FGF2, FGF5 and FGF17) or inducers of CAFs\' paracrine activity (TGF‑β1, HDGF, PDGF, CXCL8, CCL5, CCL2, IL‑6, HH and EGF) both expressed in the tumour and circulating in the blood. A total of 68 articles were selected and thoroughly analysed. The findings consistently identified upregulation of αSMA, S100A4/FSP‑1, PDGFR, PDPN, HDGF, PDGF, CXCL8, CCL5, CCL2, IL‑6, HH and EGF as poor prognostic markers in BC, while evaluation of the prognostic value of the remaining markers varied between the studies. The data confirm an association of CAFs‑specific features with BC prognosis, suggesting that both quantitative and qualitative profiling of the stroma might be required for an assessment of the true FGFR\'s clinical value.
    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
    共价药物含有可与位于蛋白质活性位点的亲核氨基酸反应的亲电子基团,特别是酶。最近,使用共价药物靶向蛋白质中的非催化氨基酸以调节困难的靶标(即靶向共价抑制剂)已经引起了相当大的兴趣。共价化合物含有多种共价反应基团(CRG),但这些CRGs中只有少数存在于FDA批准的共价药物中。
    这篇综述总结了2020-23年专利格局分析,该分析研究了围绕靶标和组织的共价药物发现领域的趋势。该分析的重点是提交给世界国际专利组织的专利申请,并根据FDA批准的药物中存在的CRG使用关键字和结构搜索的组合进行选择。
    总共确定了来自300个组织的707项专利申请,公开了在71个靶标处起作用的化合物。五个目标的专利申请计数约占总数的63%(即BTK,EGFR,FGFR,KRAS,和SARS-CoV-2Mpro)。专利数量最多的组织是一个学术机构(Dana-Farber癌症研究所)。对于一个目标,KRASG12C,新药的发现竞争激烈(>100个组织,186个专利申请)。
    UNASSIGNED: Covalent drugs contain electrophilic groups that can react with nucleophilic amino acids located in the active sites of proteins, particularly enzymes. Recently, there has been considerable interest in using covalent drugs to target non-catalytic amino acids in proteins to modulate difficult targets (i.e. targeted covalent inhibitors). Covalent compounds contain a wide variety of covalent reacting groups (CRGs), but only a few of these CRGs are present in FDA-approved covalent drugs.
    UNASSIGNED: This review summarizes a 2020-23 patent landscape analysis that examined trends in the field of covalent drug discovery around targets and organizations. The analysis focused on patent applications that were submitted to the World International Patent Organization and selected using a combination of keywords and structural searches based on CRGs present in FDA-approved drugs.
    UNASSIGNED: A total of 707 patent applications from > 300 organizations were identified, disclosing compounds that acted at 71 targets. Patent application counts for five targets accounted for ~ 63% of total counts (i.e. BTK, EGFR, FGFR, KRAS, and SARS-CoV-2 Mpro). The organization with the largest number of patent counts was an academic institution (Dana-Farber Cancer Institute). For one target, KRAS G12C, the discovery of new drugs was highly competitive (>100 organizations, 186 patent applications).
    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
    胰腺癌是一种高度致命的疾病,经常被诊断为晚期,5年总生存率约为10%。目前的治疗效果有限,强调需要新的治疗选择。本综述旨在确定和总结FGFR(成纤维细胞生长因子受体)抑制剂的临床前和临床研究,包括酪氨酸激酶抑制剂(TKIs)和FGFR特异性抑制剂,在FGFR改变的胰腺癌中。我们纳入了分析疗效的研究,安全,和不同人群的生存结果。对主要数据库的全面搜索确定了73项相关研究:32项临床前研究,16临床,和25来自灰色文学。临床试验主要集中在疗效(20项研究)和安全性(14项研究),解决生存结果的研究较少。FGFR1是研究最多的改变,其次是FGFR2和FGFR4。尽管FGFR改变在胰腺癌中相对罕见,现有数据,包括有希望的现实生活结果,提示FGFR抑制剂的巨大潜力。然而,需要更广泛的研究来确定正确的遗传驱动因素并收集可靠的生存数据。正在进行和未来的试验有望提供更全面的见解,可能导致改善FGFR改变的胰腺癌患者的靶向治疗。
    Pancreatic cancer is a highly lethal disease, often diagnosed at advanced stages, with a 5-year overall survival rate of around 10%. Current treatments have limited effectiveness, underscoring the need for new therapeutic options. This scoping review aims to identify and summarize preclinical and clinical studies on FGFR (Fibroblast Growth Factor Receptor) inhibitors, including tyrosine kinase inhibitors (TKIs) and FGFR-specific inhibitors, in pancreatic cancer with FGFR alterations. We included studies analyzing efficacy, safety, and survival outcomes in various populations. A comprehensive search across major databases identified 73 relevant studies: 32 preclinical, 16 clinical, and 25 from gray literature. The clinical trials focused primarily on efficacy (20 studies) and safety (14 studies), with fewer studies addressing survival outcomes. FGFR1 was the most studied alteration, followed by FGFR2 and FGFR4. Although FGFR alterations are relatively rare in pancreatic cancer, the available data, including promising real-life outcomes, suggest significant potential for FGFR inhibitors. However, more extensive research is needed to identify the correct genetic drivers and gather robust survival data. Ongoing and future trials are expected to provide more comprehensive insights, potentially leading to improved targeted therapies for pancreatic cancer patients with 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
    FGFR已被证明在生物过程中发挥关键作用,但它们的过度表达已被视为不同类型癌症发生和发展中的操纵子成分。出于对癌症的所有兴趣,FGFR抑制剂在过去几年中已经加速发展。因此,FGFR抑制剂是逆转耐药的主要基本工具之一,肿瘤生长,和血管生成。目前,许多FGFR抑制剂处于开发阶段或已被开发。由于巨大的需求和热点,研究人员接触了不同的药理作用,以获得结构多样的FGFR抑制剂.这里,我们选择了几个代表性的例子,如萘基,嘧啶,吡啶,吲哚,和喹啉衍生物,说明FGFR抑制剂在药物化学中的多样性和进展。这篇综述的重点是FGFR抑制剂过去五年的SAR研究,这将是一个巨大的未来范围,影响药物化学家在这一领域取得更多成就。
    FGFR have been demonstrated to perform a crucial role in biological processes but their overexpression has been perceived as the operator component in the occurrence and progression of different types of carcinoma. Out of all the interest around cancer, FGFR inhibitors have assembled pace over the past few years. Therefore, FGFR inhibitors are one of the main fundamental tools to reverse drug resistance, tumor growth, and angiogenesis. Currently, many FGFR inhibitors are under the development stage or have been developed. Due to great demand and hotspots, different pharmacophores were approached to access structurally diverse FGFR inhibitors. Here, we have selected to present several representative examples such as Naphthyl, Pyrimidine, Pyridazine, Indole, and Quinoline derivatives that illustrate the diversity and advances of FGFR inhibitors in medicinal chemistry. This review focuses on the SAR study of FGFR inhibitors last five years which will be a great future scope that influences the medicinal chemist to work towards more achievements in this area.
    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
    尽管许多努力开发用于癌症治疗的FGFR抑制剂,目前可用的FGFR抑制剂由于选择性和耐药性差引起的严重副作用,其广泛的临床应用受到了极大的限制。PROTAC技术,一种蛋白质降解的方法,与常规抑制剂相比显示出显著的优势。在我们的研究中,我们结合了Erdafitinib,泛FGFR抑制剂,用CRBN粘合剂合成和鉴定有效的FGFR2降解剂,N5.我们的研究结果表明,N5对FGFR2表现出显著的特异性和突出的酶抑制能力,对FGFR2的IC50值为0.08nM,具有很强的抗增殖活性,在0.17nM的浓度下,对胃癌细胞的抑制率保持在50%以上。机械上,N5通过降低FGFR下游蛋白水平诱导胃癌细胞周期停滞在G0/G1期和凋亡。此外,N5表现出良好的药代动力学特征,当腹膜内给药时,生物利用度为74.8%,并有效抑制SNU16异种移植肿瘤的生长,与亲本抑制剂Erdafitinib相比,表现出更高的效力。这项研究为开发和潜在应用靶向FGFR2降解的治疗剂奠定了基础。
    Despite numerous efforts to develop FGFR inhibitors for cancer treatment, the widespread clinical application of currently available FGFR inhibitors has been significantly limited due to the serious side effects caused by poor selectivity and resistance. PROTAC technology, a method for protein degradation, has shown notable advantages over conventional inhibitors. In our study, we coupled Erdafitinib, a pan-FGFR inhibitor, with a CRBN binder to synthesize and identify an effective FGFR2 degrader, N5. Our findings demonstrated that N5 displayed notable specificity for FGFR2 and outstanding enzyme inhibitory capabilities, achieving an IC50 value of 0.08 nM against FGFR2, and strong antiproliferative activity, maintaining an inhibitory rate above 50% on gastric cancer cells at a concentration of 0.17 nM. Mechanistically, N5 induced gastric cancer cell cycle arrest at the G0/G1 phase and apoptosis by decreasing the levels of FGFR downstream proteins. Moreover, N5 demonstrated favorable pharmacokinetic characteristics with a bioavailability of 74.8% when administered intraperitoneally and effectively suppressed the growth of SNU16 xenograft tumors, exhibiting greater potency compared to the parental inhibitor Erdafitinib. This study lays the groundwork for developing and potentially applying therapeutic agents targeting FGFR2 degradation.
    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
    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
    我们在健康受试者中进行了这项三部分研究,以研究tasurgratinib(成纤维细胞生长因子受体1-3的口服选择性抑制剂)和M2(其主要代谢产物)在不同条件下的药代动力学。在A部分,受试者接受tasurgratinib35mg高脂饮食喂养或禁食.在B和C部分,受试者单独接受他舒格列替尼35mg或与雷贝拉唑(酸还原剂)20mg(B部分)或利福平(CYP3A强诱导剂)600mg(C部分)一起接受治疗.主要终点是最大浓度(Cmax),和血浆浓度-时间曲线下的面积到最后可定量浓度的时间(AUC(0-t)),并外推到无限时间(AUC(0-inf))。纳入了42名受试者,14每个分为A部分,B,C.在A部分,用高脂膳食给药的他司格替尼导致了33%的Cmax和~23%的AUC(0-t)和AUC(0-inf)的下降,Cmax降低47%,M2的AUC(0-t)和AUC(0-inf)降低30%。在B部分,雷贝拉唑在稳定状态下的共同给药导致与他舒替尼的无/弱相互作用(AUC(0-t)和AUC(0-inf)增加8%,而对Cmax无影响)和M2(AUC(0-t)和AUC(0-inf)增加18%,而对Cmax无影响)。在C部分,利福平在稳定状态下的共同给药导致与他舒替尼的弱相互作用(AUC(0-t)和AUC(0-inf)降低16%)和M2(AUC(0-t)和AUC(0-inf)降低12%)。在高脂膳食的同时服用塔舒格列尼似乎减少了他舒格列尼的全身暴露,然而,与酸还原剂或CYP3A诱导剂共同给药对药代动力学的影响最小.
    We conducted this three-part study in healthy subjects to investigate the pharmacokinetics of tasurgratinib (orally available selective inhibitor of fibroblast growth factor receptor 1-3) and M2 (its major metabolite) under different conditions. In Part A, subjects received tasurgratinib 35 mg either fed with a high-fat meal or fasted. In Parts B and C, subjects received tasurgratinib 35 mg alone or with either rabeprazole (acid-reducing agent) 20 mg (Part B) or rifampin (strong CYP3A inducer) 600 mg (Part C). Primary endpoints were maximum concentration (Cmax), and areas under the plasma concentration-time curve to time of last quantifiable concentration (AUC(0-t)) and extrapolated to infinite time (AUC(0-inf)). Forty-two subjects were enrolled, 14 each into Parts A, B, and C. In Part A, administration of tasurgratinib with a high-fat meal resulted in 33% reduction in Cmax and ∼23% reduction in AUC(0-t) and AUC(0-inf) of tasurgratinib, and 47% reduction in Cmax with ∼30% reduction in AUC(0-t) and AUC(0-inf) of M2. In Part B, co-administration of rabeprazole at steady state resulted in no/weak interaction with tasurgratinib (∼8% increase in AUC(0-t) and AUC(0-inf) without an effect on Cmax) and M2 (∼18% increase in AUC(0-t) and AUC(0-inf) without an effect on Cmax). In Part C, co-administration of rifampin at steady state resulted in a weak interaction with tasurgratinib (∼16% reduction in AUC(0-t) and AUC(0-inf)) and M2 (∼12% reduction in AUC(0-t) and AUC(0-inf)). Administration of tasurgratinib with a high-fat meal appeared to reduce systemic exposure of tasurgratinib, however co-administration with an acid-reducing agent or a CYP3A inducer had a minimal impact on pharmacokinetics.
    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:血管生成因子通常在实体瘤中被激活,并且是可行的治疗靶标。然而,血管生成抑制剂(AGI)的抗癌治疗仅限于少数癌症,主要作为单一疗法,而不是根据分子适应症选择。我们旨在确定基于多分析物肿瘤询问(ETA:百科全书肿瘤分析)选择的AGI和其他抗癌剂的患者特异性组合方案是否可以扩大AGI在晚期难治性实体器官癌中的范围,并改善治疗反应。方法:我们评估了60例晚期患者的治疗结果,接受ETA指导的AGI与其他靶向联合治疗方案的难治性实体器官癌,内分泌或细胞毒性剂。治疗反应的放射学评估,然后测定客观反应率(ORR),疾病控制率(DCR),无进展生存期(PFS)和总生存期(OS)。结果:60例患者中,部分缓解(PR)28例(46.7%),29例(48.3%)观察到稳定的疾病(SD)和疾病进展(PD,在60天内)观察到3例(5.0%)。ORR为46.7%,DCR为95.0%。在最近的随访中,中位PFS(mPFS)为5.0个月,中位OS(mOS)为8.9个月。没有4级治疗相关的不良事件或治疗相关的死亡。结论:ETA指导的患者特异性联合方案与AGI和其他抗肿瘤药,与AGI单药治疗相比,可以改善预后。试验注册:所有试验的详细信息可在WHO-ICTRP:https://apps上获得。谁。int/trialsearch/。电阻IDCTRI/2018/02/011,808。液体冲击IDCTRI/2019/02/017,548。
    Background: Angiogenic factors are commonly activated in solid tumors and present a viable therapeutic target. However, anticancer treatment with angiogenesis inhibitors (AGI) is limited to a few cancers, mostly as monotherapy and not selected based on molecular indications. We aimed to determine whether patient-specific combination regimens with AGI and other anticancer agents when selected based on multi-analyte tumor interrogation (ETA: Encyclopedic Tumor Analysis) can expand the scope of AGIs in advanced refractory solid organ cancers with improved treatment responses. Methods: We evaluated treatment outcomes in 60 patients with advanced, refractory solid organ cancers who received ETA-guided combination regimens of AGI with other targeted, endocrine or cytotoxic agents. Radiological evaluation of treatment response was followed by determination of Objective Response Rate (ORR), Disease Control Rate (DCR), Progression Free Survival (PFS) and Overall Survival (OS). Results: Among the 60 patients, Partial Response (PR) was observed in 28 cases (46.7%), Stable Disease (SD) was observed in 29 cases (48.3%) and Disease Progression (PD, within 60 days) was observed in 3 cases (5.0%). The ORR was 46.7% and DCR was 95.0%. At the most recent follow-up the median PFS (mPFS) was 5.0 months and median OS (mOS) was 8.9 months. There were no Grade 4 therapy related adverse events or treatment related deaths. Conclusion: ETA-guided patient-specific combination regimens with AGI and other anti-neoplastic agents, can yield improved outcomes over AGI monotherapy. Trial Registration: Details of all trials are available at WHO-ICTRP: https://apps.who.int/trialsearch/. RESILIENT ID CTRI/2018/02/011,808. LIQUID IMPACT ID CTRI/2019/02/017,548.
    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
    自从他们在近30年前被发现以来,现在已知成纤维细胞生长因子同源因子(FHF)通过一系列机制控制可兴奋组织的功能。神经和心脏系统功能障碍是由FHF基因的功能丧失或获得突变引起的。FHF作用的最佳理解的“规范”目标是电压门控钠通道,最近的研究扩展了FHFs调节钠通道门控的方法。FHFs在可兴奋和不可兴奋细胞中的其他“非规范”功能,包括癌细胞,在过去的十几年里都有报道。这篇综述总结并评估了已报告的规范和非规范FHF功能。
    Since their discovery nearly 30 years ago, fibroblast growth factor homologous factors (FHFs) are now known to control the functionality of excitable tissues through a range of mechanisms. Nervous and cardiac system dysfunctions are caused by loss- or gain-of-function mutations in FHF genes. The best understood \'canonical\' targets for FHF action are voltage-gated sodium channels, and recent studies have expanded the repertoire of ways that FHFs modulate sodium channel gating. Additional \'non-canonical\' functions of FHFs in excitable and non-excitable cells, including cancer cells, have been reported over the past dozen years. This review summarizes and evaluates reported canonical and non-canonical FHF functions.
    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