Proto-Oncogene Proteins c-raf

原癌基因蛋白质 c - raf
  • 文章类型: Journal Article
    鼻咽癌(NPC),主要发现于中国南部地区,是一种以高度转移特性而闻名的恶性肿瘤。由远处转移和疾病复发引起的高死亡率仍然是临床上尚未解决的问题。在临床上,黄连素(BBR)化合物已广泛用于鼻咽癌治疗,以减少转移和疾病复发,并且BBR被记录为具有多种抗NPC作用的主要成分。然而,BBR抑制鼻咽癌生长和转移的机制尚不清楚。在这里,我们表明,BBR有效地抑制了生长,转移,并通过诱导特异性超级增强子(SE)入侵NPC。从机械的角度来看,RNA测序(RNA-seq)结果表明RAS-RAF1-MEK1/2-ERK1/2信号通路,由表皮生长因子受体(EGFR)激活,在BBR诱导的NPC自噬中起重要作用。自噬的阻断显著减弱了BBR介导的NPC细胞生长和转移抑制的作用。值得注意的是,BBR通过转录增加EGFR的表达,和敲除EGFR显著抑制BBR诱导的微管相关蛋白1轻链3(LC3)-II的增加和p62抑制,提示EGFR在BBR诱导的NPC自噬中起关键作用。染色质免疫沉淀测序(ChIP-seq)结果发现,仅在BBR处理的NPC细胞中存在特异性SE。这种SE敲除明显抑制了EGFR和磷酸化EGFR(EGFR-p)的表达,并逆转了BBR对NPC增殖的抑制作用。转移,和入侵。此外,BBR特异性SE可能通过增强EGFR基因转录触发自噬,从而上调RAS-RAF1-MEK1/2-ERK1/2信号通路。此外,体内BBR有效抑制NPC细胞生长和转移,随着LC3和EGFR的增加和p62的减少。总的来说,这项研究确定了一种新的BBR-特殊SE,并建立了一种新的表观遗传范式,BBR调节自噬,抑制增殖,转移,和入侵。它为BBR作为未来NPC治疗中的治疗方案的应用提供了理论基础。
    Nasopharyngeal carcinoma (NPC), primarily found in the southern region of China, is a malignant tumor known for its highly metastatic characteristics. The high mortality rates caused by the distant metastasis and disease recurrence remain unsolved clinical problems. In clinic, the berberine (BBR) compound has widely been in NPC therapy to decrease metastasis and disease recurrence, and BBR was documented as a main component with multiple anti-NPC effects. However, the mechanism by which BBR inhibits the growth and metastasis of nasopharyngeal carcinoma remains elusive. Herein, we show that BBR effectively inhibits the growth, metastasis, and invasion of NPC via inducing a specific super enhancer (SE). From a mechanistic perspective, the RNA sequencing (RNA-seq) results suggest that the RAS-RAF1-MEK1/2-ERK1/2 signaling pathway, activated by the epidermal growth factor receptor (EGFR), plays a significant role in BBR-induced autophagy in NPC. Blockading of autophagy markedly attenuated the effect of BBR-mediated NPC cell growth and metastasis inhibition. Notably, BBR increased the expression of EGFR by transcription, and knockout of EGFR significantly inhibited BBR-induced microtubule associated protein 1 light chain 3 (LC3)-II increase and p62 inhibition, proposing that EGFR plays a pivotal role in BBR-induced autophagy in NPC. Chromatin immunoprecipitation sequencing (ChIP-seq) results found that a specific SE existed only in NPC cells treated with BBR. This SE knockdown markedly repressed the expression of EGFR and phosphorylated EGFR (EGFR-p) and reversed the inhibition of BBR on NPC proliferation, metastasis, and invasion. Furthermore, BBR-specific SE may trigger autophagy by enhancing EGFR gene transcription, thereby upregulating the RAS-RAF1-MEK1/2-ERK1/2 signaling pathway. In addition, in vivo BBR effectively inhibited NPC cells growth and metastasis, following an increase LC3 and EGFR and a decrease p62. Collectively, this study identifies a novel BBR-special SE and established a new epigenetic paradigm, by which BBR regulates autophagy, inhibits proliferation, metastasis, and invasion. It provides a rationale for BBR application as the treatment regime in NPC therapy in 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
    RAS/RAF/MEK/ERK信号的异常激活对于KIT突变介导的胃肠道间质瘤(GIST)的肿瘤发生至关重要。在这项研究中,我们发现抑制RAF1抑制了GIST中野生型KIT和原发性KIT突变的激活,原发性KIT突变显示出更高的敏感性。这表明KIT和RAF1之间存在正反馈回路,其中RAF1促进KIT信号传导。我们进一步证明RAF1与KIT相关,RAF1的激酶活性对于其对KIT激活的贡献是必需的。因此,抑制RAF1抑制细胞存活,扩散,以及由野生型KIT和原发性KIT突变介导的体外细胞周期进程。体内抑制RAF1抑制了携带种系KIT/V558A突变的转基因小鼠模型中的GIST生长,显示与伊马替尼相似的治疗效率,GIST的一线靶向治疗药物,而联合使用伊马替尼和RAF1抑制剂进一步抑制了肿瘤的生长。获得KIT的耐药次级突变是GIST靶向治疗后治疗失败的主要原因。像野生型KIT和原发性KIT突变一样,抑制RAF1抑制了继发性KIT突变的激活,和细胞存活,扩散,体外细胞周期进程,和继发性KIT突变介导的体内肿瘤生长。然而,与原发性KIT突变相比,继发性KIT突变的激活对RAF1的依赖性较小.一起来看,我们的结果表明,RAF1促进KIT信号和KIT突变介导的GIST的肿瘤发生,提供进一步研究RAF1抑制剂单独或与KIT抑制剂联合治疗GIST的理由,特别是在对KIT抑制剂有抗性的情况下。
    The aberrant activation of RAS/RAF/MEK/ERK signaling is important for KIT mutation-mediated tumorigenesis of gastrointestinal stromal tumor (GIST). In this study, we found that inhibition of RAF1 suppresses the activation of both wild-type KIT and primary KIT mutations in GIST, with primary KIT mutations showing greater sensitivity. This suggests a positive feedback loop between KIT and RAF1, wherein RAF1 facilitates KIT signaling. We further demonstrated that RAF1 associates with KIT and the kinase activity of RAF1 is necessary for its contribution to KIT activation. Accordingly, inhibition of RAF1 suppressed cell survival, proliferation, and cell cycle progression in vitro mediated by both wild-type KIT and primary KIT mutations. Inhibition of RAF1 in vivo suppressed GIST growth in a transgenic mouse model carrying germline KIT/V558A mutation, showing a similar treatment efficiency as imatinib, the first-line targeted therapeutic drug of GIST, while the combination use of imatinib and RAF1 inhibitor further suppressed tumor growth. Acquisition of drug-resistant secondary mutation of KIT is a major cause of treatment failure of GIST following targeted therapy. Like wild-type KIT and primary KIT mutations, inhibition of RAF1 suppressed the activation of secondary KIT mutation, and the cell survival, proliferation, cell cycle progression in vitro, and tumor growth in vivo mediated by secondary KIT mutation. However, the activation of secondary KIT mutation is less dependent on RAF1 compared with that of primary KIT mutations. Taken together, our results revealed that RAF1 facilitates KIT signaling and KIT mutation-mediated tumorigenesis of GIST, providing a rationale for further investigation into the use of RAF1 inhibitors alone or in combination with KIT inhibitor in the treatment of GIST, particularly in cases resistant to KIT 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
    RAF激酶是通过RAS-RAF-MEK-ERK途径进行信号转导所必需的,它们的活性在人类癌症和放射病发育综合征中经常上调。由于其复杂的激活过程,开发有效靶向RAF功能的药物一直是一项具有挑战性的工作,强调需要更详细地了解英国皇家空军的规定。这篇综述将侧重于最近的结构和生化研究,这些研究为英国皇家空军的监管周期提供了“快照”。揭示自抑制BRAF单体的结构,活性BRAF和CRAF同源二聚体,以及含有CRAF或BRAFV600E的HSP90/CDC37伴侣复合物。此外,我们将描述有关BRAF如何在其调节状态之间转变的见解,并研究各种BRAF结构域和14-3-3二聚体在维持BRAF作为自抑制单体和促进其向活性二聚体转变方面的作用。我们还将讨论HSP90/CDC37伴侣复合物在稳定CRAF和某些致癌BRAF突变体的蛋白质水平方面的功能,并作为PP5蛋白磷酸酶介导的RAF去磷酸化的平台。最后,我们将讨论BRAF和CRAF之间观察到的监管差异,以及这些差异如何影响BRAF和CRAF作为人类疾病驱动因素的功能。
    The RAF kinases are required for signal transduction through the RAS-RAF-MEK-ERK pathway, and their activity is frequently up-regulated in human cancer and the RASopathy developmental syndromes. Due to their complex activation process, developing drugs that effectively target RAF function has been a challenging endeavor, highlighting the need for a more detailed understanding of RAF regulation. This review will focus on recent structural and biochemical studies that have provided \'snapshots\' into the RAF regulatory cycle, revealing structures of the autoinhibited BRAF monomer, active BRAF and CRAF homodimers, as well as HSP90/CDC37 chaperone complexes containing CRAF or BRAFV600E. In addition, we will describe the insights obtained regarding how BRAF transitions between its regulatory states and examine the roles that various BRAF domains and 14-3-3 dimers play in both maintaining BRAF as an autoinhibited monomer and in facilitating its transition to an active dimer. We will also address the function of the HSP90/CDC37 chaperone complex in stabilizing the protein levels of CRAF and certain oncogenic BRAF mutants, and in serving as a platform for RAF dephosphorylation mediated by the PP5 protein phosphatase. Finally, we will discuss the regulatory differences observed between BRAF and CRAF and how these differences impact the function of BRAF and CRAF as drivers of human disease.
    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
    婴儿纤维肉瘤(IFS)和先天性中胚层肾瘤(CMN)是婴儿期和儿童早期罕见的肌纤维母细胞肿瘤,通常具有ETV6::NTRK3基因融合。IFS/CMN被认为是具有“中间预后”的肿瘤,因为它们具有局部侵袭性,但很少转移,通常会有一个有利的结果。一部分IFS/CMN相关肿瘤对ETV6::NTRK3基因重排呈阴性,其特征在于其他嵌合蛋白促进MAPK信号上调。在这些肿瘤的很大一部分中,被分类为IFS样间质肿瘤,有贡献的分子事件仍有待鉴定。这里,我们报告了8个ETV6::NTRK3基因融合阴性肿瘤中涉及RAF1的三种不同的重排,其组织学诊断为IFS/CMN.这三种融合蛋白保留了激酶的整个催化结构域。两种嵌合产品,GOLGA4::RAF1和LRRFIP2::RAF1以前曾被报道为不同癌症的驱动事件,而第三个,CLIP1::RAF1代表一种新的融合蛋白。我们证明CLIP1::RAF1作为一种真正的癌蛋白,通过MAPK信号的组成性上调促进细胞增殖和迁移。我们证明CLIP1::RAF1过度活跃行为不需要RAS激活,并且是由嵌合蛋白的组成型14-3-3蛋白非依赖性二聚化介导的。正如之前报道的ETV6::NTRK3融合蛋白,CLIP1::RAF1类似地上调PI3K-AKT信号传导。我们的发现记录了RAF1基因重排代表ETV6::NTRK3阴性IFS/CMN的复发事件,并为在这些癌症中使用抑制剂抑制MAPK和PI3K-AKT信号提供了理论基础。©2024英国和爱尔兰病理学会。
    Infantile fibrosarcomas (IFS) and congenital mesoblastic nephroma (CMN) are rare myofibroblastic tumors of infancy and early childhood commonly harboring the ETV6::NTRK3 gene fusion. IFS/CMN are considered as tumors with an \'intermediate prognosis\' as they are locally aggressive, but rarely metastasize, and generally have a favorable outcome. A fraction of IFS/CMN-related neoplasms are negative for the ETV6::NTRK3 gene rearrangement and are characterized by other chimeric proteins promoting MAPK signaling upregulation. In a large proportion of these tumors, which are classified as IFS-like mesenchymal neoplasms, the contributing molecular events remain to be identified. Here, we report three distinct rearrangements involving RAF1 among eight ETV6::NTRK3 gene fusion-negative tumors with an original histological diagnosis of IFS/CMN. The three fusion proteins retain the entire catalytic domain of the kinase. Two chimeric products, GOLGA4::RAF1 and LRRFIP2::RAF1, had previously been reported as driver events in different cancers, whereas the third, CLIP1::RAF1, represents a novel fusion protein. We demonstrate that CLIP1::RAF1 acts as a bona fide oncoprotein promoting cell proliferation and migration through constitutive upregulation of MAPK signaling. We show that the CLIP1::RAF1 hyperactive behavior does not require RAS activation and is mediated by constitutive 14-3-3 protein-independent dimerization of the chimeric protein. As previously reported for the ETV6::NTRK3 fusion protein, CLIP1::RAF1 similarly upregulates PI3K-AKT signaling. Our findings document that RAF1 gene rearrangements represent a recurrent event in ETV6::NTRK3-negative IFS/CMN and provide a rationale for the use of inhibitors directed to suppress MAPK and PI3K-AKT signaling in these cancers. © 2024 The Pathological Society of Great Britain and Ireland.
    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
    RAS/丝裂原活化蛋白激酶(MAPK)途径的功能障碍是人类癌症的常见驱动因素。因此,既是通路的主调节器,RAS,和它的近端激酶效应,RAF,几十年来一直是人们关注的药物靶标。重要的是,RAS/MAPK通路内的信号是高度协调的,由于形成称为RAS/RAF信号体的高阶复合物,它可能最低限度地含有RAS和RAF前体的二聚体。在疾病状态下,RAS和RAF以同源和/或异二聚体形式组装。传统上,RAS和RAF的药物开发活动都利用了纯化重组蛋白的生化测定。由于这些测定不查询RAS或RAF蛋白质在细胞中的全长和复合形式,使用这些测定收集的效力结果通常不能与MAPK途径的抑制相关联。为了更准确地量化此信号组件的参与,我们提出了一种基于生物发光共振能量转移(BRET)的方法,以有条件地测量活细胞中RAS/RAF信号小体中单个质子的靶标接合。
    Dysfunction of the RAS/mitogen-activated protein kinase (MAPK) pathway is a common driver of human cancers. As such, both the master regulator of the pathway, RAS, and its proximal kinase effectors, RAFs, have been of interest as drug targets for decades. Importantly, signaling within the RAS/MAPK pathway is highly coordinated due to the formation of a higher-order complex called the RAS/RAF signalosome, which may minimally contain dimers of both RAS and RAF protomers. In the disease state, RAS and RAF assemble in homo- and/or heterodimeric forms. Traditionally, drug development campaigns for both RAS and RAF have utilized biochemical assays of purified recombinant protein. As these assays do not query the RAS or RAF proteins in their full-length and complexed forms in cells, potency results collected using these assays have often failed to correlate with inhibition of the MAPK pathway. To more accurately quantify engagement at this signaling components, we present a bioluminescence resonance energy transfer (BRET)-based method to conditionally measure target engagement at individual protomers within the RAS/RAF signalosome in live cells.
    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:东亚黑色素瘤发病率呈上升趋势,然而,在这个人群中缺乏分子景观的研究。我们检查了在韩国一个三级中心接受下一代测序(NGS)的黑色素瘤患者,专注于接受belvarafenib的有NRAS或RAF改变的患者,一种泛RAF二聚体抑制剂,通过扩展访问计划(EAP)。
    方法:收集了在2017年11月至2023年5月期间接受NGS的192例黑色素瘤患者的数据。获得并注释了变体调用格式数据。EAP患者每天两次接受450mg的belvarafenib。
    结果:RAS/RTK途径的改变是最普遍的,BRAF和NRAS的变化率分别为22.4%和17.7%,分别。NGS能够额外检测融合突变,包括6个BRAF和1个RAF1融合。16例NRAS或RAF改变的患者通过EAP接受了belvarafenib,观察到50%的疾病控制,2名患者表现出显著的反应。
    结论:我们的研究强调了NGS在检测BRAF中的价值,NRAS突变和RAF融合,扩大恶性黑色素瘤靶向治疗的可能性。Belvarafenib在具有这些改变的患者中显示出临床益处。正在进行的试验将为belvarafenib的安全性和有效性提供进一步的见解。
    BACKGROUND: Melanoma incidence is on the rise in East Asia, yet studies of the molecular landscape are lacking in this population. We examined patients with melanoma who underwent next-generation sequencing (NGS) at a single tertiary center in South Korea, focusing on patients harboring NRAS or RAF alterations who received belvarafenib, a pan-RAF dimer inhibitor, through the Expanded Access Program (EAP).
    METHODS: Data were collected from 192 patients with melanoma who underwent NGS between November 2017 and May 2023. Variant call format data were obtained and annotated. Patients in the EAP received 450 mg twice daily doses of belvarafenib.
    RESULTS: Alterations in the RAS/RTK pathway were the most prevalent, with BRAF and NRAS alteration rates of 22.4% and 17.7%, respectively. NGS enabled additional detection of fusion mutations, including 6 BRAF and 1 RAF1 fusion. Sixteen patients with NRAS or RAF alterations received belvarafenib through the EAP, and disease control was observed in 50%, with 2 patients demonstrating remarkable responses.
    CONCLUSIONS: Our study highlights the value of NGS in detecting BRAF, NRAS mutations and RAF fusions, expanding possibilities for targeted therapies in malignant melanoma. Belvarafenib showed clinical benefit in patients harboring these alterations. Ongoing trials will provide further insights into the safety and efficacy of belvarafenib.
    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
    胰腺癌在肿瘤学中代表了一个巨大的挑战,主要是由于其侵略性和有限的治疗选择。胰腺导管腺癌(PDAC)患者的预后,胰腺癌的主要形式,令人失望的是,5年总生存率仅为5%。几乎95%的PDAC患者携带Kirsten大鼠肉瘤病毒(KRAS)致癌突变。KRAS激活下游细胞内通路,最值得注意的是快速加速的纤维肉瘤(RAF)/丝裂原活化蛋白激酶激酶(MEK)/细胞外信号调节激酶(ERK)信号轴。RAF/MEK/ERK通路的失调是胰腺癌的一个重要特征,因此它的主要组成部分,英国皇家空军,MEK和ERK激酶,已经在药理学上成为目标,主要是小分子抑制剂。近年来抑制剂的发展不仅直接靶向RAF/MEK/ERK通路,而且通过抑制其调节因子间接靶向RAF/MEK/ERK通路,例如含Src同源性的蛋白酪氨酸磷酸酶2(SHP2)和七同源物1(SOS1),提供新的治疗机会。此外,针对突变KRAS变异体的等位基因特异性小分子抑制剂的发现为在对抗胰腺癌的斗争中的成功创新带来了兴奋.在这里,我们回顾了靶向治疗和组合策略的最新进展,重点是当前的临床前和临床方法,提供关键的见解,强调这些努力的潜力,并支持他们改善PDAC患者生活的承诺。
    Pancreatic cancer represents a formidable challenge in oncology, primarily due to its aggressive nature and limited therapeutic options. The prognosis of patients with pancreatic ductal adenocarcinoma (PDAC), the main form of pancreatic cancer, remains disappointingly poor with a 5-year overall survival of only 5%. Almost 95% of PDAC patients harbor Kirsten rat sarcoma virus (KRAS) oncogenic mutations. KRAS activates downstream intracellular pathways, most notably the rapidly accelerated fibrosarcoma (RAF)/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling axis. Dysregulation of the RAF/MEK/ERK pathway is a crucial feature of pancreatic cancer and therefore its main components, RAF, MEK and ERK kinases, have been targeted pharmacologically, largely by small-molecule inhibitors. The recent advances in the development of inhibitors not only directly targeting the RAF/MEK/ERK pathway but also indirectly through inhibition of its regulators, such as Src homology-containing protein tyrosine phosphatase 2 (SHP2) and Son of sevenless homolog 1 (SOS1), provide new therapeutic opportunities. Moreover, the discovery of allele-specific small-molecule inhibitors against mutant KRAS variants has brought excitement for successful innovations in the battle against pancreatic cancer. Herein, we review the recent advances in targeted therapy and combinatorial strategies with focus on the current preclinical and clinical approaches, providing critical insight, underscoring the potential of these efforts and supporting their promise to improve the lives of patients with PDAC.
    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
    RAF蛋白激酶是MAPK通路的重要效应子,是重要的肿瘤药物靶点。到目前为止,对RAF激活的结构理解是基于低温电子显微镜(cryo-EM)和BRAF在不同构象状态下的X射线结构,作为与KRAS的非活性或活性复合物,14-3-3和MEK1。在这项研究中,我们已经使用CRAF激酶结构域在昆虫细胞中表达为组成型活性Y340D/Y341D突变体,解决了分辨率为3.4µ的CRAF2/14-3-32和分辨率为4.2µ的CRAF2/14-3-32/MEK12的第一个低温EM结构。我们的CRAF2/14-3-32和CRAF2/14-3-32/MEK12低温-EM结构的整体结构与与14-3-3或14-3-3/MEK1复合的相应BRAF结构高度相似,并代表活化的二聚体RAF构象。我们的CRAF低温EM结构为激活的CRAF2/14-3-32/MEK12复合物的结构理解提供了额外的见解。
    RAF protein kinases are essential effectors in the MAPK pathway and are important cancer drug targets. Structural understanding of RAF activation is so far based on cryo-electron microscopy (cryo-EM) and X-ray structures of BRAF in different conformational states as inactive or active complexes with KRAS, 14-3-3 and MEK1. In this study, we have solved the first cryo-EM structures of CRAF2/14-3-32 at 3.4 Å resolution and CRAF2/14-3-32/MEK12 at 4.2 Å resolution using CRAF kinase domain expressed as constitutively active Y340D/Y341D mutant in insect cells. The overall architecture of our CRAF2/14-3-32 and CRAF2/14-3-32/MEK12 cryo-EM structures is highly similar to corresponding BRAF structures in complex with 14-3-3 or 14-3-3/MEK1 and represent the activated dimeric RAF conformation. Our CRAF cryo-EM structures provide additional insights into structural understanding of the activated CRAF2/14-3-32/MEK12 complex.
    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
    暂无摘要。
    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
    RAS/丝裂原激活的蛋白激酶(MAPK)信号级联在人类恶性肿瘤中通常通过RAS或RAF癌基因驱动的过程失调。在RAF激酶家族的成员中,CRAF在RAS-MAPK信号通路中发挥重要作用,以及癌症的进展。最近的研究提供了证据,表明CRAF在生理调节中的作用以及通过MAPK依赖性和MAPK非依赖性机制对BRAF抑制剂的抗性。然而,仅靶向CRAF激酶活性的有效性仍存在争议.此外,CRAF的激酶非依赖性功能对于KRAS突变的肺癌可能是必需的.必须制定策略以增强RAS或RAF癌基因驱动的肿瘤的功效并使毒性最小化。该综述调查了在癌症中观察到的CRAF改变,并揭示了CRAF在由多种癌基因推动的癌症中的独特作用。这篇综述还旨在总结CRAF相互作用蛋白,并描述CRAF在各种癌症标志中的调控。此外,我们讨论了pan-RAF抑制剂的最新进展及其与其他治疗方法的联合应用,以改善RAF/RAS突变肿瘤患者的治疗结果并最大限度地减少不良反应.通过全面了解CRAF在癌症中的多方面作用,并强调RAF抑制剂治疗的最新进展,我们努力确定协同靶标并阐明抗性途径,为更稳健和更安全的癌症治疗组合策略奠定了基础。
    The RAS/mitogen-activated protein kinase (MAPK) signaling cascade is commonly dysregulated in human malignancies by processes driven by RAS or RAF oncogenes. Among the members of the RAF kinase family, CRAF plays an important role in the RAS-MAPK signaling pathway, as well as in the progression of cancer. Recent research has provided evidence implicating the role of CRAF in the physiological regulation and the resistance to BRAF inhibitors through MAPK-dependent and MAPK-independent mechanisms. Nevertheless, the effectiveness of solely targeting CRAF kinase activity remains controversial. Moreover, the kinase-independent function of CRAF may be essential for lung cancers with KRAS mutations. It is imperative to develop strategies to enhance efficacy and minimize toxicity in tumors driven by RAS or RAF oncogenes. The review investigates CRAF alterations observed in cancers and unravels the distinct roles of CRAF in cancers propelled by diverse oncogenes. This review also seeks to summarize CRAF-interacting proteins and delineate CRAF\'s regulation across various cancer hallmarks. Additionally, we discuss recent advances in pan-RAF inhibitors and their combination with other therapeutic approaches to improve treatment outcomes and minimize adverse effects in patients with RAF/RAS-mutant tumors. By providing a comprehensive understanding of the multifaceted role of CRAF in cancers and highlighting the latest developments in RAF inhibitor therapies, we endeavor to identify synergistic targets and elucidate resistance pathways, setting the stage for more robust and safer combination strategies for cancer treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号