KRAS inhibitor

  • 文章类型: Journal Article
    背景:多药耐药(MDR)限制了成功的癌症化疗。P-糖蛋白(P-gp),BCRP和MRP1是MDR的关键触发因素。不幸的是,迄今为止,FDA尚未批准MDR调节剂.这里,我们将研究BI-2865,一种泛KRAS抑制剂,逆转P-gp诱导的MDR,BCRP和MRP1在体外和体内,及其逆转机制将被探索。
    方法:通过MTT法检测BI-2865的细胞毒性及其体外MDR去除效果,并通过P-gp介导的小鼠KBv200异种移植物评估体内相应的逆转功能。通过荧光多柔比星流式细胞术实验估计BI-2865诱导的细胞内药物释放和保留的改变,并通过LC-MS分析异种移植物肿瘤中的化疗药物积累。BI-2865抑制P-gp底物流出的机制通过钒酸敏感的ATP酶测定进行分析,[125I]-IAAP-光标记测定和计算机分子对接。通过蛋白质印迹法检测BI-2865对P-gp表达和KRAS下游信号的影响。流式细胞术和/或qRT-PCR。通过免疫荧光观察P-gp的亚细胞定位。
    结果:我们发现BI-2865显著增强了P-gp驱动的MDR癌细胞对包括紫杉醇在内的化疗药物的反应,长春新碱和阿霉素,而在其亲代敏感细胞和BCRP或MRP1驱动的MDR细胞中未观察到这种作用。重要的是,小鼠体内联合研究证实,BI-2865可有效提高紫杉醇的抗肿瘤作用,且无毒性损伤。在机制上,BI-2865通过直接阻断P-gp的外排功能促使阿霉素在癌细胞中积累,更具体地说,通过BI-2865竞争性结合P-gp的药物结合位点来实现。更重要的是,在有效的MDR逆转浓度下,BI-2865既不改变P-gp的表达和位置,也不降低其下游AKT或ERK1/2信号活性。
    结论:本研究揭示了BI-2865作为MDR调节剂的新应用,这可能被用来有效地,安全和特异性地提高临床P-gp介导的MDR难治性癌症的化疗疗效。
    BACKGROUND: Multidrug resistance (MDR) limits successful cancer chemotherapy. P-glycoprotein (P-gp), BCRP and MRP1 are the key triggers of MDR. Unfortunately, no MDR modulator was approved by FDA to date. Here, we will investigate the effect of BI-2865, a pan-KRAS inhibitor, on reversing MDR induced by P-gp, BCRP and MRP1 in vitro and in vivo, and its reversal mechanisms will be explored.
    METHODS: The cytotoxicity of BI-2865 and its MDR removal effect in vitro were tested by MTT assays, and the corresponding reversal function in vivo was assessed through the P-gp mediated KBv200 xenografts in mice. BI-2865 induced alterations of drug discharge and reservation in cells were estimated by experiments of Flow cytometry with fluorescent doxorubicin, and the chemo-drug accumulation in xenografts\' tumor were analyzed through LC-MS. Mechanisms of BI-2865 inhibiting P-gp substrate\'s efflux were analyzed through the vanadate-sensitive ATPase assay, [125I]-IAAP-photolabeling assay and computer molecular docking. The effects of BI-2865 on P-gp expression and KRAS-downstream signaling were detected via Western blotting, Flow cytometry and/or qRT-PCR. Subcellular localization of P-gp was visualized by Immunofluorescence.
    RESULTS: We found BI-2865 notably fortified response of P-gp-driven MDR cancer cells to the administration of chemo-drugs including paclitaxel, vincristine and doxorubicin, while such an effect was not observed in their parental sensitive cells and BCRP or MRP1-driven MDR cells. Importantly, the mice vivo combination study has verified that BI-2865 effectively improved the anti-tumor action of paclitaxel without toxic injury. In mechanism, BI-2865 prompted doxorubicin accumulating in carcinoma cells by directly blocking the efflux function of P-gp, which more specifically, was achieved by BI-2865 competitively binding to the drug-binding sites of P-gp. What\'s more, at the effective MDR reversal concentrations, BI-2865 neither varied the expression and location of P-gp nor reduced its downstream AKT or ERK1/2 signaling activity.
    CONCLUSIONS: This study uncovered a new application of BI-2865 as a MDR modulator, which might be used to effectively, safely and specifically improve chemotherapeutic efficacy in the clinical P-gp mediated MDR refractory 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: Case Reports
    一名KirstenRas(KRAS)G12C突变的非小细胞肺癌(NSCLC)患者表现不佳,并通过一线KRAS靶向治疗sotorasib获得混合反应。疾病进展后,化疗加免疫疗法获得部分缓解。NSCLC中KRASG12C突变的免疫环境可能有利于免疫治疗。
    KRAS是最常见的突变基因之一,过去是不可能的。II期CodeBreak100试验显示,在先前接受KRAS抑制剂治疗的KRASG12C突变NSCLC患者中,有6.8个月的中位无进展生存期(PFS)和12.5个月的总生存期(OS)。Sotorasib.大脑的标本,淋巴结(LN),和患者的血液通过下一代测序进行分析。进行苏木精和伊红染色和免疫组织化学以进行病理表征。计算机断层扫描(CT)和磁共振成像(MRI)扫描用于治疗反应评估。该患者被诊断为东部肿瘤协作组(ECOG-PS)表现不佳,患有转移性KRASG12C突变的肺腺癌,其对索托拉西作为一线治疗的混合反应。虽然5个月的PFS与sotorasib治疗并不奇怪,患者ECOG-PS评分从4分显著提高至1分.随后,培美曲塞联合派姆单抗治疗实现部分缓解(PR).此病例突出了索托拉西一线治疗对未经治疗的KRASG12C突变患者的优异疗效。化疗加免疫疗法的高疗效表明,KRASG12C突变患者的免疫环境可能有利于免疫疗法。
    UNASSIGNED: One Kirsten Ras (KRAS) G12C mutated non-small cell lung cancer (NSCLC) patient had improved poor performance status and obtained mixed response with the first-line KRAS-targeted treatment of sotorasib. After disease progression, partial response was achieved with chemotherapy plus immunotherapy. KRAS G12C mutated immunoenvironment in NSCLC may favor the immunotherapy.
    UNASSIGNED: KRAS is one of the most commonly mutated genes, which used to be untargetable. The phase II CodeBreak 100 trial revealed 6.8-month median progress-free survival (PFS) and 12.5-month overall survival (OS) in previously treated KRAS G12C-mutant NSCLC patients treated with KRAS inhibitor, sotorasib. The specimens of the brain, lymph node (LN), and blood from the patient were analyzed by next-generation sequencing. Hematoxylin and eosin staining and immunohistochemistry were performed for pathological characterization. Computed tomography (CT) and magnetic resonance imaging (MRI) scan were used for treatment response evaluation. The patient was diagnosed in a bad Eastern Cooperative Oncology Group performance status (ECOG-PS) with metastatic KRAS G12C-mutated lung adenocarcinoma who had achieved mixed response to sotorasib as the first-line treatment. Although 5-month PFS of the treatment with sotorasib was not surprising, the patient achieved significantly improved ECOG-PS score from 4 to 1. Subsequently, partial response (PR) was achieved with the treatment of pemetrexed plus pembrolizumab. This case highlights superior efficacy of first-line treatment with sotorasib for the advance untreated KRAS G12C-mutant patients. The high efficacy of the treatment with chemotherapy plus immunotherapy revealed that immunoenvironment of KRAS G12C-mutated patient may favor the immunotherapy.
    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
    KRASG12C抑制剂(adagrasib和sotorasib)已显示出靶向KRASG12C突变肺癌的临床前景;然而,大多数患者最终会产生抵抗力。在KRASG12C和STK11/LKB1共突变的腺癌患者中,我们发现,在治疗前活检中,鳞状细胞癌基因特征的富集与对adagrasib的应答不良相关.Lkb1缺陷的KRASG12C和KrasG12D肺癌小鼠模型和用KRAS抑制剂治疗的类器官的研究揭示了肿瘤调用谱系可塑性程序。腺-鳞状过渡(AST),能够抵抗KRAS抑制。转录组和表观基因组分析揭示ΔNp63驱动AST并调节对KRAS抑制的反应。我们鉴定了由AST可塑性特征和Krt6a的表达标记的中等高塑性细胞状态。值得注意的是,基线时AST可塑性特征和KRT6A的表达与不良的adagrasib反应相关。这些数据表明AST在KRAS抑制剂抗性中的作用,并为肺癌中的KRAS靶向疗法提供预测性生物标志物。
    KRASG12C inhibitors (adagrasib and sotorasib) have shown clinical promise in targeting KRASG12C-mutated lung cancers; however, most patients eventually develop resistance. In lung patients with adenocarcinoma with KRASG12C and STK11/LKB1 co-mutations, we find an enrichment of the squamous cell carcinoma gene signature in pre-treatment biopsies correlates with a poor response to adagrasib. Studies of Lkb1-deficient KRASG12C and KrasG12D lung cancer mouse models and organoids treated with KRAS inhibitors reveal tumors invoke a lineage plasticity program, adeno-to-squamous transition (AST), that enables resistance to KRAS inhibition. Transcriptomic and epigenomic analyses reveal ΔNp63 drives AST and modulates response to KRAS inhibition. We identify an intermediate high-plastic cell state marked by expression of an AST plasticity signature and Krt6a. Notably, expression of the AST plasticity signature and KRT6A at baseline correlates with poor adagrasib responses. These data indicate the role of AST in KRAS inhibitor resistance and provide predictive biomarkers for KRAS-targeted therapies in lung cancer.
    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大鼠肉瘤病毒癌基因(KRAS),第一个发现的人类癌基因,长期以来一直被认为是“不可吸毒”。KRAS突变经常发生在多种人类癌症中,包括非小细胞肺癌(NSCLC)。结直肠癌(CRC)和胰腺导管腺癌(PDAC),充当“分子开关”,决定各种致癌信号通路的激活。除了其内在的促肿瘤作用,KRAS改变还表现出独特的免疫特征,其特征在于PD-L1水平升高和高肿瘤突变负荷(TMB)。KRAS突变通过阻止有效的T细胞浸润和招募抑制性免疫细胞(包括骨髓来源的抑制细胞(MDSC))来塑造免疫抑制微环境,调节性T细胞(Tregs),癌相关成纤维细胞(CAFs)。在免疫检查点抑制剂(ICI)时代,与具有完整KRAS的患者相比,具有突变KRAS的NSCLC患者倾向于对ICI更敏感。KRAS突变的标志是存在多种共突变。不同类型的共同改变具有不同的肿瘤微环境(TME)特征和对ICI的响应。TP53共突变具有“热”TME,对免疫疗法具有更高的反应,而其他功能丧失突变与“较冷”TME和基于ICI的治疗的不良结果相关。KRASG12C抑制剂的突破性发现显着改善了这种KRAS亚型的预后,尽管疗效仅限于NSCLC患者。KRASG12C抑制剂也能恢复抑制性TME,创造与ICI组合的机会。然而,KRAS抑制剂的一个不可避免的挑战是耐药性。有希望的组合策略,如与SHP2的组合,是一种值得进一步探索的方法,因为它们的免疫调节作用。
    Kirsten rats sarcoma viral oncogene (KRAS), the first discovered human oncogene, has long been recognized as \"undruggable\". KRAS mutations frequently occur in multiple human cancers including non-small cell lung cancer(NSCLC), colorectal cancer(CRC) and pancreatic ductal adenocarcinoma(PDAC), functioning as a \"molecule switch\" determining the activation of various oncogenic signaling pathways. Except for its intrinsic pro-tumorigenic role, KRAS alteration also exhibits an unique immune signature characterized by elevated PD-L1 level and high tumor mutational burden(TMB). KRAS mutation shape an immune suppressive microenvironment by impeding effective T cells infiltration and recruiting suppressive immune cells including myeloid-derived suppressor cells(MDSCs), regulatory T cells(Tregs), cancer associated fibroblasts(CAFs). In immune checkpoint inhibitor(ICI) era, NSCLC patients with mutated KRAS tend to be more responsive to ICI than patients with intact KRAS. The hallmark for KRAS mutation is the existence of multiple kinds of co-mutations. Different types of co-alterations have distinct tumor microenvironment(TME) signatures and responses to ICI. TP53 co-mutation possess a \"hot\" TME and achieve higher response to immunotherapy while other loss of function mutation correlated with a \"colder\" TME and a poor outcome to ICI-based therapy. The groundbreaking discovery of KRAS G12C inhibitors significantly improved outcomes for this KRAS subtype even though efficacy was limited to NSCLC patients. KRAS G12C inhibitors also restore the suppressive TME, creating an opportunity for combinations with ICI. However, an inevitable challenge to KRAS inhibitors is drug resistance. Promising combination strategies such as combination with SHP2 is an approach deserve further exploration because of their immune modulatory effect.
    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