KRAS G12D

  • 文章类型: Journal Article
    KRASG12D是实体瘤中最常见的突变的致癌KRAS亚型,在临床上仍然无法用药。这里,我们发展了很高的亲和力,选择性,长效,和非共价KRASG12D抑制剂,HRS-4642,具有0.083nM的亲和常数。HRS-4642在体外和体内均表现出对KRASG12D突变癌症的强大功效。重要的是,在一期临床试验中,HRS-4642在逐步增加的给药队列中表现出有希望的抗肿瘤活性。此外,通过全基因组CRISPR-Cas9筛选,破译了HRS-4642的致敏和抗性谱,这揭示了蛋白酶体作为一个敏感的目标。我们进一步观察到蛋白酶体抑制剂,Carfilzomib,提高了HRS-4642的抗肿瘤疗效。此外,HRS-4642,作为单一药物或与卡非佐米联合使用,将肿瘤微环境重塑为免疫许可。总之,这项研究为KRASG12D突变癌症患者提供了潜在的治疗方法,目前缺乏有效的治疗方法。
    KRAS G12D is the most frequently mutated oncogenic KRAS subtype in solid tumors and remains undruggable in clinical settings. Here, we developed a high affinity, selective, long-acting, and non-covalent KRAS G12D inhibitor, HRS-4642, with an affinity constant of 0.083 nM. HRS-4642 demonstrated robust efficacy against KRAS G12D-mutant cancers both in vitro and in vivo. Importantly, in a phase 1 clinical trial, HRS-4642 exhibited promising anti-tumor activity in the escalating dosing cohorts. Furthermore, the sensitization and resistance spectrum for HRS-4642 was deciphered through genome-wide CRISPR-Cas9 screening, which unveiled proteasome as a sensitization target. We further observed that the proteasome inhibitor, carfilzomib, improved the anti-tumor efficacy of HRS-4642. Additionally, HRS-4642, either as a single agent or in combination with carfilzomib, reshaped the tumor microenvironment toward an immune-permissive one. In summary, this study provides potential therapies for patients with KRAS G12D-mutant cancers, for whom effective treatments are currently lacking.
    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
    KRASG12D是几种癌症中最常见的致癌突变。因此,针对KRASG12D的抑制剂设计代表了一种有希望的抗癌治疗策略.MRTX1133是KRASG12D的高效抑制剂(近似实验Kd≈0.0002nM),目前处于1/2期研究,然而,化合物与KRASG12D结合的途径仍然未知,复杂的动态过程背后的机制是具有挑战性的实验捕获,这阻碍了基于结构的抗癌药物设计。这里,使用MRTX1133作为探针,无偏分子动力学(MD)用于模拟MRTX1133自发结合KRASG12D的过程。在42个独立MD模拟中的6个(总共99μs),观察到MRTX1133与KRASG12D成功结合。动力学亚稳态是指通过马尔可夫状态模型(MSM)分析揭示了MRTX1133与KRASG12D结合的潜在途径。此外,通过MM/GBSA分析鉴定了对于MRTX1133识别和在优选的低能量状态下的紧密结合所必需的8个关键残基。总之,本研究为理解MRTX1133与KRASG12D结合的途径和机制提供了新的视角。
    KRAS G12D is the most common oncogenic mutation identified in several types of cancer. Therefore, design of inhibitors targeting KRAS G12D represents a promising strategy for anticancer therapy. MRTX1133 is a highly potent inhibitor (approximate experiment Kd ≈ 0.0002 nM) of KRAS G12D and is currently in Phase 1/2 study, however, pathways of the compound binding to KRAS G12D has remained unknown, and the mechanism underlying the complicated dynamic process are challenging to capture experimentally, which hinder the structure-based anti-cancer drug design. Here, using MRTX1133 as a probe, unbiased molecular dynamics (MD) was used to simulate the process of MRTX1133 spontaneously binding to KRAS G12D. In six of 42 independent MD simulation (a total of 99 μs), MRTX1133 was observed to successfully associate with KRAS G12D. The kinetically metastable states refer to the potential pathways of MRTX1133 binding to KRAS G12D were revealed by Markov state models (MSM) analysis. Additionally, 8 key residues that are essential for MRTX1133 recognition and tight binding at the preferred low energy states were identified by MM/GBSA analysis. In sum, this study provides a new perspective on understanding the pathways and mechanism of MRTX1133 binding to KRAS G12D.
    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
    Kirsten大鼠肉瘤病毒癌蛋白同源物(KRAS)目前是肿瘤学家和转化科学家的主要关注点,由KRAS靶向治疗非小细胞肺癌(NSCLC)患者的令人兴奋的结果驱动。虽然KRAS突变继续导致高癌症诊断和死亡,研究人员开发了针对KRAS变异的独特策略。在过去的40年中进行了调查,由于缺乏药物结合袋,被认为是“不可吸毒”,最近的突破和加速FDA批准的第一个共价抑制剂靶向KRASG12C,在很大程度上引发了进一步的药物开发。小分子开发已经靶向先前鉴定的原发性位置改变,如G12、G13、Q61,并且扩展以解决新出现的继发性突变和获得性抗性。感兴趣的,非共价KRASG12D靶向抑制剂MRTX-1133在人源化胰腺癌小鼠模型中显示了有希望的结果,并且似乎正在从实验室到床边.虽然该手稿正在审查中,但已发表了一类针对G12D的新型共价抑制剂,这些所谓的麦芽糖内酯可以交联G12D的GDP和GTP结合形式。后一种状态的抑制在体外和小鼠异种移植物中抑制下游信号传导和癌细胞增殖。此外,非共价泛KRAS抑制剂,BI-2865,降低细胞系和小鼠模型中的肿瘤增殖。最后,下一代KRAS突变体特异性和泛RAS三复合物抑制剂彻底改变了RAS药物的发现。这篇综述将通过新出现的次级突变和获得性抗性的镜头,对当前一代的KRAS抑制剂进行结构生物学研究。
    The Kirsten rat sarcoma viral oncoprotein homolog (KRAS) is currently a primary focus of oncologists and translational scientists, driven by exciting results with KRAS-targeted therapies for non-small cell lung cancer (NSCLC) patients. While KRAS mutations continue to drive high cancer diagnosis and death, researchers have developed unique strategies to target KRAS variations. Having been investigated over the past 40 years and considered \"undruggable\" due to the lack of pharmacological binding pockets, recent breakthroughs and accelerated FDA approval of the first covalent inhibitors targeting KRASG12C, have largely sparked further drug development. Small molecule development has targeted the previously identified primary location alterations such as G12, G13, Q61, and expanded to address the emerging secondary mutations and acquired resistance. Of interest, the non-covalent KRASG12D targeting inhibitor MRTX-1133 has shown promising results in humanized pancreatic cancer mouse models and is seemingly making its way from bench to bedside. While this manuscript was under review a novel class of first covalent inhibitors specific for G12D was published, These so-called malolactones can crosslink both GDP and GTP bound forms of G12D. Inhibition of the latter state suppressed downstream signaling and cancer cell proliferation in vitro and in mouse xenografts. Moreover, a non-covalent pan-KRAS inhibitor, BI-2865, reduced tumor proliferation in cell lines and mouse models. Finally, the next generation of KRAS mutant-specific and pan-RAS tri-complex inhibitors have revolutionized RAS drug discovery. This review will give a structural biology perspective on the current generation of KRAS inhibitors through the lens of emerging secondary mutations and acquired 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
    Kirsten大鼠肉瘤(KRAS)病毒原癌基因的单点突变是人类癌症的最常见原因。在人类中,致癌KRAS突变负责约30%的肺,胰腺,和结肠癌。主要突变KRASG12D变体之一是胰腺癌的原因,是一个有吸引力的药物靶标。在撰写本文时,没有食品和药物管理局(FDA)批准的药物可用于KRASG12D突变体.所以,有必要开发一种有效的KRASG12D药物。寻找新药的过程昂贵且耗时。另一方面,计算机药物设计方法具有成本效益且耗时较少。在这里,我们采用了机器学习算法,如K最近邻(KNN),支持向量机(SVM),和随机森林(RF)用于鉴定针对KRASG12D突变体的新抑制剂。预测总共82个命中对KRASG12D突变体有活性。将活性命中对接到KRASG12D突变体的活性位点。此外,为了评估具有良好对接评分的化合物的稳定性,对前两个复合物和标准复合物(MRTX-1133)进行200nsMD模拟。与标准化合物相比,前两个命中显示出高稳定性。与标准化合物相比,前两个命中的结合能是良好的。我们确定的命中具有抑制KRASG12D突变的潜力,可以帮助对抗癌症。据我们所知,这是第一项基于机器学习的虚拟筛选的研究,分子对接,并进行了分子动力学模拟,以鉴定KRASG12D突变体的新抑制剂。
    Single-point mutations in the Kirsten rat sarcoma (KRAS) viral proto-oncogene are the most common cause of human cancer. In humans, oncogenic KRAS mutations are responsible for about 30% of lung, pancreatic, and colon cancers. One of the predominant mutant KRAS G12D variants is responsible for pancreatic cancer and is an attractive drug target. At the time of writing, no Food and Drug Administration (FDA) approved drugs are available for the KRAS G12D mutant. So, there is a need to develop an effective drug for KRAS G12D. The process of finding new drugs is expensive and time-consuming. On the other hand, in silico drug designing methodologies are cost-effective and less time-consuming. Herein, we employed machine learning algorithms such as K-nearest neighbor (KNN), support vector machine (SVM), and random forest (RF) for the identification of new inhibitors against the KRAS G12D mutant. A total of 82 hits were predicted as active against the KRAS G12D mutant. The active hits were docked into the active site of the KRAS G12D mutant. Furthermore, to evaluate the stability of the compounds with a good docking score, the top two complexes and the standard complex (MRTX-1133) were subjected to 200 ns MD simulation. The top two hits revealed high stability as compared to the standard compound. The binding energy of the top two hits was good as compared to the standard compound. Our identified hits have the potential to inhibit the KRAS G12D mutation and can help combat cancer. To the best of our knowledge, this is the first study in which machine-learning-based virtual screening, molecular docking, and molecular dynamics simulation were carried out for the identification of new promising inhibitors for the KRAS G12D mutant.
    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
    致癌RAS突变,通常在人类肿瘤中观察到,影响大约30%的癌症病例,并对有效的癌症治疗构成重大挑战。目前抑制KRASG12D突变的策略显示出有限的成功,强调迫切需要新的治疗方法。在这项研究中,我们设计并合成了几种嘌呤和嘧啶类似物作为KRASG12D突变的抑制剂。我们合成的化合物对具有KRASG12D突变的细胞系表现出有效的抗癌活性,有效地阻碍了他们的成长。它们在正常细胞中也表现出低毒性,表明它们对携带KRASG12D突变的癌细胞具有选择性作用。值得注意的是,铅化合物,PU1-1诱导KRASG12D突变细胞的程序性细胞死亡,并降低活性KRAS及其下游信号蛋白的水平。此外,PU1-1在KRASG12D突变诱导的胰腺异种移植模型中显著缩小肿瘤大小,进一步验证其作为治疗剂的潜力。这些发现强调了基于嘌呤的KRASG12D抑制剂作为靶向癌症治疗候选药物的潜力。然而,这些化合物的进一步探索和优化对于满足KRAS突变癌症患者未满足的临床需求至关重要.
    Oncogenic RAS mutations, commonly observed in human tumors, affect approximately 30% of cancer cases and pose a significant challenge for effective cancer treatment. Current strategies to inhibit the KRAS G12D mutation have shown limited success, emphasizing the urgent need for new therapeutic approaches. In this study, we designed and synthesized several purine and pyrimidine analogs as inhibitors for the KRAS G12D mutation. Our synthesized compounds demonstrated potent anticancer activity against cell lines with the KRAS G12D mutation, effectively impeding their growth. They also exhibited low toxicity in normal cells, indicating their selective action against cancer cells harboring the KRAS G12D mutation. Notably, the lead compound, PU1-1 induced the programmed cell death of KRAS G12D-mutated cells and reduced the levels of active KRAS and its downstream signaling proteins. Moreover, PU1-1 significantly shrunk the tumor size in a pancreatic xenograft model induced by the KRAS G12D mutation, further validating its potential as a therapeutic agent. These findings highlight the potential of purine-based KRAS G12D inhibitors as candidates for targeted cancer therapy. However, further exploration and optimization of these compounds are essential to meet the unmet clinical needs of patients with KRAS-mutant cancers.
    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:KRAS突变是NSCLC的常见驱动因素,肺癌患者中KRASG12C和G12D突变的比例较高。KRAS以前被认为是“不可下药”的目标,但首款KRASG12C突变靶向药物AMG510,于2021年进入市场。然而,G12D突变肿瘤的治疗仍有待发现。丹酚酸F(SalF),一种来自中药丹参(SM)的单体,和KRAS有很高的结合亲和力,特别是KRASG12D.迫切需要研究针对KRASG12D驱动的NSCLC的有效和安全的新型靶向疗法。
    方法:为了评估SalF的抗癌作用,我们在体外使用过表达KRAS的肺癌细胞,皮下移植肿瘤模型,和KRASG12D小鼠体内模型。然后,使用分子对接研究了SalF和KRAS的结合作用,蛋白水解测定和蛋白质热移位测定。更关键的是,我们利用RT-qPCR和Western印迹法研究了肺中PI3K/AKT信号通路.
    结果:这是首次评估SalF对体内KRAS过表达的肺癌细胞或KRASG12D肺肿瘤的抗癌作用的研究。我们证明SalF抑制OE-KRASA549细胞迁移,体外增殖和促进细胞凋亡。此外,我们使用皮下移植肿瘤模型显示SalF在体内抑制肺癌细胞的生长。有趣的是,我们小组发现SalF与G12D强结合,可以通过分子对接降低稳定性并促进KRASG12D突变体的降解,蛋白水解测定和蛋白质热移位测定。进一步的研究表明,在KrasG12D小鼠模型中,经过SalF治疗,小鼠肺部肿瘤的数量和大小显著减少。更重要的是,SalF可通过抑制下游PI3K/AKT信号通路激活促进细胞凋亡。
    结论:SalF激活细胞凋亡信号通路,抑制抗凋亡基因,抑制肺癌细胞生长。这些数据表明,SalF可以有效抑制KRASG12D突变的肺肿瘤的生长。SalF可能是一种新型的KRASG12D抑制剂,KRAS突变肺癌的临床治疗提供了有力的理论依据。
    BACKGROUND: KRAS mutation is a common driver of NSCLC, and there is a high proportion of lung cancer patients with KRAS G12C and G12D mutation. KRAS was previously considered an \"undruggable\" target, but the first KRAS G12C mutation-targeted drug AMG510, entered the market in 2021. However, treatments for G12D mutant tumors remain to be discovered. Salvianolic acid F (SalF), a monomer derived from the traditional Chinese medicine Salvia miltiorrhiza (SM), and KRAS had high binding affinity, especially for KRAS G12D. There is an urgent need to investigate effective and safe novel targeted therapies against KRAS G12D-driven NSCLC.
    METHODS: To evaluate the anticancer effect of SalF, we used KRAS-overexpressing lung cancer cells in vitro, a subcutaneous transplant tumor model, and KRAS G12D mice model in vivo. Then, the binding effect of SalF and KRAS was investigated using molecular docking, proteolytic assays and protein thermal shift assays. More critically, the PI3K/AKT signaling pathway in the lung was investigated utilizing RT-qPCR and Western Blotting.
    RESULTS: This is the first study to evaluate the anticancer effect of SalF on KRAS-overexpressing lung cancer cells or KRAS G12D lung tumors in vivo. We demonstrated that SalF inhibits OE-KRAS A549 cell migration, proliferation and promotes apoptosis in vitro. In addition, we used a subcutaneous transplant tumor model to show that SalF suppresses the growth of lung cancer cells in vivo. Interestingly, our group found that SalF was strongly bound to G12D and could decrease the stability and promoted the degradation of the KRAS G12D mutant through molecular docking, proteolytic assays and protein thermal shift assays. Further research demonstrated that in the KrasG12D mice model, after SalF treatment, the number and size of mouse lung tumors were significantly reduced. More importantly, SalF can promote apoptosis by inhibiting downstream PI3K/AKT signaling pathway activation.
    CONCLUSIONS: SalF activated apoptosis signaling pathways, suppressed anti-apoptotic genes, and inhibited lung cancer cell growth. These datas suggested that SalF could effectively inhibit the growth of lung tumors with KRAS G12D mutation. SalF may be a novel inhibitor against KRAS G12D, providing a strong theoretical basis for the clinical treatment of lung cancer with KRAS mutations.
    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
    突变KRAS诱导的肿瘤发生高度参与胰腺的进展,肺,和乳腺癌。相对而言,KRASG12D和KRASG12C是促进癌症进展和侵袭性的最常见突变。尽管KRAS突变抑制剂具有显著的治疗潜力,日复一日,他们在患者中变得耐药。基于多表位的癌症疫苗是诱导针对肿瘤抗原的免疫应答的有希望的替代策略。在本研究中,我们设计了,constructed,并使用反向疫苗学和免疫信息学方法验证了针对KRASG12D和G12C突变体的新型多表位疫苗构建体。此外,疫苗构建体在结构上经过精炼,并显示出显著的生理化学性质,并能诱导免疫反应.此外,通过计算机克隆将优化的疫苗构建体克隆到pET‑28a(+)表达载体中。最后,多表位疫苗构建体结构稳定,可溶性,抗原性,非过敏,无毒。Further,它必须在体外和体内进行研究,以评估其在不久的将来对KRAS突变癌症的治疗效果。
    Mutant KRAS-induced tumorigenesis is highly involved in the progression of pancreatic, lung, and breast cancer. Comparatively, KRAS G12D and KRAS G12C are the most frequent mutations that promote cancer progression and aggressiveness. Although KRAS mutant inhibitors exhibit significant therapeutic potential, day by day, they are becoming resistant among patients. Multi-epitope based cancer vaccines are a promising alternative strategy that induces an immune response against tumor antigens. In the present study, we have designed, constructed, and validated a novel multi-epitope vaccine construct against KRAS G12D and G12C mutants using reverse vaccinology and immunoinformatics approaches. In addition, the vaccine construct was structurally refined and showed significant physiochemical properties, and could induce an immune response. Furthermore, the optimized vaccine construct was cloned into a pET‑28a (+) expression vector through in silico cloning. Conclusively, the multi-epitope vaccine construct is structurally stable, soluble, antigenic, non‑allergic, and non‑toxic. Further, it has to be studied in in vitro and in vivo to evaluate its therapeutic efficacy against KRAS-mutated cancers in the near future.
    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
    KRAS是癌症中最常见的突变癌基因。激活密码子12的突变,尤其是G12D,在一系列癌和腺癌中患病率最高。随着KRAS-G12D抑制剂进入临床试验,了解KRAS-G12D癌症的生物学,识别预测治疗反应的生物标志物至关重要。在这篇评论中,我们讨论了KRAS-G12D腺癌的基因组学和生物学,包括组织学特征,转录景观,免疫微环境,以及这些因素如何影响对治疗的反应。此外,我们探索使用新型G12D抑制剂的潜在治疗策略,利用从使用G12C抑制剂的临床试验中获得的知识。
    KRAS is the most frequently mutated oncogene in cancer. Activating mutations in codon 12, especially G12D, have the highest prevalence across a range of carcinomas and adenocarcinomas. With inhibitors to KRAS-G12D now entering clinical trials, understanding the biology of KRAS-G12D cancers, and identifying biomarkers that predict therapeutic response is crucial. In this Review, we discuss the genomics and biology of KRAS-G12D adenocarcinomas, including histological features, transcriptional landscape, the immune microenvironment, and how these factors influence response to therapy. Moreover, we explore potential therapeutic strategies using novel G12D inhibitors, leveraging knowledge gained from clinical trials using G12C 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
    像KRAS这样的“不可用”目标在药物开发中尤其具有挑战性。我们设计了一种新颖的化学击倒策略,CANDDY(具有亲和力的化学击倒Nd降解动力学)技术,其使用与从蛋白酶体抑制剂修饰的降解标签(CANDDY标签)缀合的小分子(CANDDY分子)促进蛋白质降解。我们证明了CANDDY标签允许直接蛋白酶体靶标降解而不依赖于泛素化。我们合成了一种KRAS降解CANDDY分子,TUS-007,其在KRAS突变体(G12D和G12V)和野生型KRAS中诱导降解。我们证实了TUS-007在腹膜内给药的人结肠细胞皮下异种移植模型(KRASG12V)和口服给药的人胰腺细胞原位异种移植模型(KRASG12D)中的肿瘤抑制作用。因此,CANDDY技术有可能在治疗上靶向以前不可用的蛋白质,提供了一种更简单、更实用的药物靶向方法,并避免了E3酶与靶标匹配的困难。
    \"Undruggable\" targets such as KRAS are particularly challenging in the development of drugs. We devised a novel chemical knockdown strategy, CANDDY (Chemical knockdown with Affinity aNd Degradation DYnamics) technology, which promotes protein degradation using small molecules (CANDDY molecules) that are conjugated to a degradation tag (CANDDY tag) modified from proteasome inhibitors. We demonstrated that CANDDY tags allowed for direct proteasomal target degradation independent of ubiquitination. We synthesized a KRAS-degrading CANDDY molecule, TUS-007, which induced degradation in KRAS mutants (G12D and G12V) and wild-type KRAS. We confirmed the tumor suppression effect of TUS-007 in subcutaneous xenograft models of human colon cells (KRAS G12V) with intraperitoneal administrations and in orthotopic xenograft models of human pancreatic cells (KRAS G12D) with oral administrations. Thus, CANDDY technology has the potential to therapeutically target previously undruggable proteins, providing a simpler and more practical drug targeting approach and avoiding the difficulties in matchmaking between the E3 enzyme and the target.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Multicenter Study
    目的:Kirsten大鼠肉瘤病毒(KRAS)癌基因的突变构成了肺腺癌的重要驱动因素,存在于10-40%的患者中,表现出异质性的临床结果,主要由并发遗传改变驱动。然而,在拉丁美洲,KRAS突变亚型的特征及其对临床结局的影响有限.
    方法:在墨西哥国家癌症研究所(INCan)进行了一项队列研究。腺癌和KRAS突变提前分期的个体,通过下一代测序检测,接受至少一行治疗的患者被纳入分析.从2014年6月至2023年3月的机构数据库中检索临床和病理特征。
    结果:346例患者中有54例(15.6%)发现了KRAS,其中50例纳入分析。KRASG12D(n=16,32%)和KRASG12C(n=16,32%)代表最普遍的亚型。KRASG12D突变与女性相关(p=0.018),从不吸烟者(p=0.108),并与EGFR(25.0%vs.17.6%,p=0.124)和CDKN2A(18.8%与14.7%,p=0.157)。KRASG12D患者表现出更好的ORR(66.6%vs.30.0%;OR4.66,95%CI1.23-17.60,p=0.023),多变量分析与较好的PFS(HR0.36,95%CI0.16-0.80;p=0.012)和OS(HR0.24,95%CI0.08-0.70;p=0.009)显着相关。
    结论:据我们所知,这项研究首次全面描述了拉丁美洲患者KRAS突变型NSCLC的分子异质性.我们的数据加强了目前的观点,即KRAS突变的NSCLC不是单一的癌基因驱动的疾病,并强调了在这个基因组定义的NSCLC子集中不同分子谱的预后影响。在更大的多中心拉丁美洲队列中需要进一步验证,以证实我们的发现。
    OBJECTIVE: Mutations in the Kirsten rat sarcoma viral (KRAS) oncogene constitute a significant driver of lung adenocarcinoma, present in 10-40% of patients, which exhibit heterogeneous clinical outcomes, mainly driven by concurrent genetic alterations. However, characterization of KRAS mutational subtypes and their impact on clinical outcomes in Latin America is limited.
    METHODS: A cohort study was conducted at the National Cancer Institute (INCan) of Mexico. Individuals with advance-staged of adenocarcinoma and KRAS mutations, detected by next-generation sequencing, having undergone at least one line of therapy were included for analysis. Clinical and pathological characteristics were retrieved from institutional database from June 2014 to March 2023.
    RESULTS: KRAS was identified in fifty-four (15.6%) of 346 patients, among which 50 cases were included for analysis. KRASG12D (n = 16, 32%) and KRASG12C (n = 16, 32%) represented the most prevalent subtypes. KRASG12D mutations were associated with female (p = 0.018), never smokers (p = 0.108), and concurrences with EGFR (25.0% vs. 17.6%, p = 0.124) and CDKN2A (18.8% vs. 14.7%, p = 0.157). KRASG12D patients showed a better ORR (66.6% vs. 30.0%; OR 4.66, 95% CI 1.23-17.60, p = 0.023) and on multivariate analysis was significantly associated with better PFS (HR 0.36, 95% CI 0.16-0.80; p = 0.012) and OS (HR 0.24, 95% CI 0.08-0.70; p = 0.009).
    CONCLUSIONS: To our knowledge, this study represents the first effort to comprehensively characterize the molecular heterogeneity of KRAS-mutant NSCLC in Latin American patients. Our data reinforce the current view that KRAS-mutated NSCLC is not a single oncogene-driven disease and emphasizes the prognostic impact of diverse molecular profiles in this genomically defined subset of NSCLC. Further validation is warranted in larger multicenter Latin American cohorts to confirm our findings.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号