HER3

HER3
  • 文章类型: Journal Article
    转移性乳腺癌(mBC)对女性健康构成重大威胁,是女性恶性肿瘤的主要原因。人表皮生长因子受体(HER)3,ErbB/HER受体酪氨酸激酶家族的重要成员,是磷酸肌醇-3激酶/蛋白激酶B信号通路的关键激活剂。HER3过表达显著促进对靶向其他HER受体的药物的抗性的发展,如HER2和表皮生长因子受体,在mBC的发生和发展中起着至关重要的作用。最近,许多HER3靶向治疗剂,如单克隆抗体(mAb),双特异性抗体(bAbs),和抗体-药物缀合物(ADC),出现了。然而,单独使用时,HER3靶向的单克隆抗体和单克隆抗体的疗效有限,它们的组合可能导致毒性副作用。另一方面,ADC对癌细胞有细胞毒性,可以通过抗体与靶细胞结合,这突出了它们在mBC的靶向HER3治疗中的用途。这篇综述概述了HER3研究的最新进展,历史性举措,和靶向HER3治疗转移性乳腺癌的创新方法。评估当前方法的优缺点可能会产生有价值的见解和教训。
    Metastatic breast cancer (mBC) poses a significant threat to women\'s health and is a major cause of malignant neoplasms in women. Human epidermal growth factor receptor (HER)3, an integral member of the ErbB/HER receptor tyrosine kinase family, is a crucial activator of the phosphoinositide-3 kinase/protein kinase B signaling pathway. HER3 overexpression significantly contributes to the development of resistance to drugs targeting other HER receptors, such as HER2 and epidermal growth factor receptors, and plays a crucial role in the onset and progression of mBC. Recently, numerous HER3-targeted therapeutic agents, such as monoclonal antibodies (mAbs), bispecific antibodies (bAbs), and antibody-drug conjugates (ADCs), have emerged. However, the efficacy of HER3-targeted mAbs and bAbs is limited when used individually, and their combination may result in toxic adverse effects. On the other hand, ADCs are cytotoxic to cancer cells and can bind to target cells through antibodies, which highlights their use in targeted HER3 therapy for mBC. This review provides an overview of recent advancements in HER3 research, historical initiatives, and innovative approaches in targeted HER3 therapy for metastatic breast cancer. Evaluating the advantages and disadvantages of current methods may yield valuable insights and lessons.
    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
    人表皮生长因子受体3(HER3),是她家庭的一部分,在各种人类癌症中异常表达。由于HER3仅具有弱的酪氨酸激酶活性,当HER3配体神经调节素1(NRG1)或神经调节素2(NRG2)出现时,活化的HER3通过与其他受体形成异二聚体,有助于癌症发展和耐药性,主要包括表皮生长因子受体(EGFR)和人表皮生长因子受体2(HER2)。抑制HER3及其下游信号,包括PI3K/AKT,MEK/MAPK,JAK/STAT,和Src激酶,被认为是克服耐药性和提高治疗效率的必要条件。直到现在,尽管多种抗HER3抗体正在进行临床前和临床研究,由于HER3靶向疗法的安全性和有效性,因此均未获得用于临床癌症治疗的许可.因此,开发具有安全性的HER3靶向药物,耐受性,敏感性对临床癌症治疗至关重要。本文就HER3耐药机制的研究进展作一综述,在临床前和临床试验中进行的HER3靶向治疗,以及一些可用作HER3未来设计药物的新兴分子,旨在为未来针对HER3的抗癌药物的研发提供见解。
    Human epidermal growth factor receptor 3 (HER3), which is part of the HER family, is aberrantly expressed in various human cancers. Since HER3 only has weak tyrosine kinase activity, when HER3 ligand neuregulin 1 (NRG1) or neuregulin 2 (NRG2) appears, activated HER3 contributes to cancer development and drug resistance by forming heterodimers with other receptors, mainly including epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2). Inhibition of HER3 and its downstream signaling, including PI3K/AKT, MEK/MAPK, JAK/STAT, and Src kinase, is believed to be necessary to conquer drug resistance and improve treatment efficiency. Until now, despite multiple anti-HER3 antibodies undergoing preclinical and clinical studies, none of the HER3-targeted therapies are licensed for utilization in clinical cancer treatment because of their safety and efficacy. Therefore, the development of HER3-targeted drugs possessing safety, tolerability, and sensitivity is crucial for clinical cancer treatment. This review summarizes the progress of the mechanism of HER3 in drug resistance, the HER3-targeted therapies that are conducted in preclinical and clinical trials, and some emerging molecules that could be used as future designed drugs for HER3, aiming to provide insights for future research and development of anticancer drugs targeting HER3.
    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:HER3(ErbB3),人类表皮生长因子受体家族的成员,在各种癌症中经常过度表达。多种HER3靶向抗体和抗体-药物缀合物(ADC)被开发用于实体瘤治疗。然而,目前还没有一种HER3靶向药物被批准用于肿瘤治疗.我们开发了DB-1310,一种HER3ADC,由一种新型人源化抗HER3单克隆抗体与专有DNA拓扑异构酶I抑制剂有效载荷(P1021)共价连接组成,并评估DB-1310在临床前模型中的疗效和安全性。
    方法:通过ELISA和SPR测量DB-1310与Her3和其他HER家族的结合。通过FACS测试DB-1310和帕特单抗的结合表位的竞争。乳房的敏感性,肺,通过体外细胞杀伤试验评估了DB-1310的前列腺癌和结肠癌细胞系。体内生长抑制研究评估了DB-1310对Her3+乳腺的敏感性,肺,结肠癌和前列腺癌异种移植模型。还在食蟹猴中测量了安全性特征。
    结果:DB-1310通过具有高亲和力和内化能力的新型表位结合HER3。体外,DB-1310在许多HER3+乳腺中表现出细胞毒性,肺,前列腺癌和结肠癌细胞系。HER3+HCC1569乳腺癌的体内研究,NCI-H441肺癌和Colo205结肠癌异种移植模型显示DB-1310具有剂量依赖性杀肿瘤活性。在HER3+非小细胞肺癌(NSCLC)和前列腺癌患者来源的异种移植物(PDX)模型中也观察到肿瘤抑制。此外,DB-1310显示出比派妥克替康(HER3-DXd)更强的肿瘤生长抑制活性,这是在相同剂量下临床开发中的另一种HER3ADC。DB-1310的抑瘤活性与EGFR酪氨酸激酶抑制剂协同作用,奥希替尼,并在奥希替尼耐药的PDX模型中发挥作用。食蟹猴的安全性的临床前评估进一步显示,DB-1310具有良好的安全性,最高非严重毒性剂量(HNSTD)为45mg/kg。
    结论:这些发现表明,DB-1310在体外和体内模型中对HER3+肿瘤具有有效的抗肿瘤活性,并在非临床物种中显示出可接受的安全性。因此,DB-1310可有效用于HER3+实体瘤的临床治疗。
    BACKGROUND: HER3 (ErbB3), a member of the human epidermal growth factor receptor family, is frequently overexpressed in various cancers. Multiple HER3-targeting antibodies and antibody-drug conjugates (ADCs) were developed for the solid tumor treatment, however none of HER3-targeting agent has been approved for tumor therapy yet. We developed DB-1310, a HER3 ADC composed of a novel humanized anti-HER3 monoclonal antibody covalently linked to a proprietary DNA topoisomerase I inhibitor payload (P1021), and evaluate the efficacy and safety of DB-1310 in preclinical models.
    METHODS: The binding of DB-1310 to Her3 and other HER families were measured by ELISA and SPR. The competition of binding epitope for DB-1310 and patritumab was tested by FACS. The sensitivity of breast, lung, prostate and colon cancer cell lines to DB-1310 was evaluated by in vitro cell killing assay. In vivo growth inhibition study evaluated the sensitivity of DB-1310 to Her3 + breast, lung, colon and prostate cancer xenograft models. The safety profile was also measured in cynomolgus monkey.
    RESULTS: DB-1310 binds HER3 via a novel epitope with high affinity and internalization capacity. In vitro, DB-1310 exhibited cytotoxicity in numerous HER3 + breast, lung, prostate and colon cancer cell lines. In vivo studies in HER3 + HCC1569 breast cancer, NCI-H441 lung cancer and Colo205 colon cancer xenograft models showed DB-1310 to have dose-dependent tumoricidal activity. Tumor suppression was also observed in HER3 + non-small cell lung cancer (NSCLC) and prostate cancer patient-derived xenograft (PDX) models. Moreover, DB-1310 showed stronger tumor growth-inhibitory activity than patritumab deruxtecan (HER3-DXd), which is another HER3 ADC in clinical development at the same dose. The tumor-suppressive activity of DB-1310 synergized with that of EGFR tyrosine kinase inhibitor, osimertinib, and exerted efficacy also in osimertinib-resistant PDX model. The preclinical assessment of safety in cynomolgus monkeys further revealed DB-1310 to have a good safety profile with a highest non severely toxic dose (HNSTD) of 45 mg/kg.
    CONCLUSIONS: These finding demonstrated that DB-1310 exerted potent antitumor activities against HER3 + tumors in in vitro and in vivo models, and showed acceptable safety profiles in nonclinical species. Therefore, DB-1310 may be effective for the clinical treatment of HER3 + solid tumors.
    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
    抗体-药物缀合物(ADC)-用于靶向肿瘤治疗的一类突破性药物-由具有强抗原特异性的单克隆抗体与高活性细胞毒性剂(也称为“有效载荷”)组成。在过去的20年里,乳腺癌研究已经发展成为ADC研究和开发的焦点,最近出版了几本具有里程碑意义的出版物。这些进步正在开创乳腺癌治疗的变革时代,并通过引入称为“HER2-low”的前瞻性亚型来重新定义常规分类。“ADC的最新迭代显示,通过优化各种因素,提高了疾病管理的功效,特别是纳入旁观者效应。这些缀合物不再限于致癌驱动人表皮生长因子受体2(HER2)。其他抗原,包括人类表皮生长因子受体3(HER3),滋养层细胞表面抗原2(Trop-2),锌转运蛋白ZIP6(LIV-1),和叶酸受体α(FRα),最近出现了有趣的肿瘤细胞表面非驱动基因靶标的ADC,每个都有一个或多个特定的ADC,在乳腺癌领域显示出令人鼓舞的结果。本文就近年来ADCs在HER2-low乳腺癌治疗中的应用进展作一综述。此外,本综述探讨了影响靶点选择对ADC疗效影响的潜在因素,为优化晚期乳腺癌HER2低表达个体ADC的临床应用提供新的见解.
    Antibody-drug conjugates (ADCs)-a groundbreaking class of agents for targeted oncological therapies-consist of monoclonal antibodies with strong antigenic specificity coupled with highly active cytotoxic agents (also referred to as \"payloads\"). Over the past 2 decades, breast cancer research has evolved into a focal point for the research and development of ADCs, leading to several recent landmark publications. These advancements are ushering in a transformative era in breast cancer treatment and redefining conventional classifications by introducing a prospective subtype termed \"HER2-low.\" The latest iterations of ADCs have demonstrated enhanced efficacy in disease management through the optimization of various factors, notably the incorporation of the bystander effect. These conjugates are no longer limited to the oncogenic driver human epidermal growth factor receptor 2 (HER2). Other antigens, including human epidermal growth factor receptor 3 (HER3), trophoblast cell surface antigen 2 (Trop-2), zinc transporter ZIP6 (LIV-1), and folate receptor α (FRα), have recently emerged as intriguing tumor cell surface nondriver gene targets for ADCs, each with one or more specific ADCs that showed encouraging results in the breast cancer field. This article reviews recent advances in the application of ADCs in the treatment of HER2-low breast cancer. Additionally, this review explores the underlying factors contributing to the impact of target selection on ADC efficacy to provide new insights for optimizing the clinical application of ADCs in individuals with low HER2 expression in advanced breast 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
    HER3(人表皮生长因子受体3)在各种癌症中经常过表达,包括非小细胞肺癌(NSCLC),原发性肿瘤的患病率为83%。它参与肿瘤发生和对靶向治疗的抗性使HER3成为癌症治疗的有希望的靶标。尽管由于缺乏催化活性而最初被认为“不可用”,抗HER3疗法的开发取得了重大进展。单克隆抗体如lumretuzumab,seribantumab,和帕特单抗已经显示出靶向HER3克服表皮生长因子受体(EGFR)酪氨酸激酶抑制剂(TKIs)耐药性的潜力。此外,抗体-药物偶联物(ADC)如HER3-DXd(patritumabderuxtecan)是新的候选药物,已证明通过利用HER3的广泛表达将细胞毒性化学物质选择性递送至NSCLC细胞,最小化细胞毒性。这篇综述旨在评估当前HER3治疗的疗效及其在非小细胞肺癌中的治疗潜力。纳入临床试验的证据。
    HER3 (Human Epidermal Growth Factor Receptor 3) is frequently overexpressed in various cancers, including non-small cell lung cancer (NSCLC), with a prevalence of 83% in primary tumors. Its involvement in tumorigenesis and resistance to targeted therapies makes HER3 a promising target for cancer treatment. Despite being initially considered \"undruggable\" due to its lack of catalytic activity, significant progress has been made in the development of anti-HER3 therapeutics. Monoclonal antibodies such as lumretuzumab, seribantumab, and patritumab have shown potential in targeting HER3 to overcome resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs). Additionally, antibody-drug conjugates (ADCs) like HER3-DXd (patritumab deruxtecan) are new drug candidates that have demonstrated selective delivery of cytotoxic chemicals to NSCLC cells by exploiting HER3\'s widespread expression, minimizing cytotoxicity. This review aims to evaluate the efficacy of current HER3 therapeutics in development and their therapeutic potential in NSCLC, incorporating evidence from 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
    HER3(人表皮生长因子受体3)通过与EGFR(表皮生长因子受体)或HER2的异源二聚化作用,在激活磷酸肌醇3激酶(PI3K)和AKT信号传导中起重要作用,这是促进肿瘤细胞存活的关键途径。HER3是癌症治疗的一个有希望的靶点,一些针对HER3的抗体已经进入临床试验.在这项研究中,我们表征了一种新型的抗HER3单克隆抗体,SIBP-03.SIBP-03(0.01-10μg/mL)特异性和浓度依赖性地阻断了神经调节素(NRG)依赖性和非依赖性HER3激活,减弱HER3介导的下游信号并抑制细胞增殖。这种抗肿瘤活性是依赖性的,至少在某种程度上,关于SIBP-03诱导的,细胞介导的细胞毒性和细胞吞噬作用。重要的是,SIBP-03在体外和体内增强EGFR或HER2靶向药物(西妥昔单抗或曲妥珠单抗)的抗肿瘤活性。这种协同作用的潜在机制涉及HER3介导的下游信号传导的抑制增加。总的来说,这些结果表明,目前正在中国进行I期临床试验的SIBP-03,可能为携带激活HER3的癌症患者提供新的治疗选择,特别是作为组合治疗策略的一部分.
    HER3 (human epidermal growth factor receptor 3) acts through heterodimerization with EGFR (epidermal growth factor receptor) or HER2 to play an essential role in activating phosphoinositide 3-kinase (PI3K) and AKT signaling-a crucial pathway that promotes tumor cell survival. HER3 is a promising target for cancer therapy, and several HER3-directed antibodies have already entered into clinical trials. In this study we characterized a novel anti-HER3 monoclonal antibody, SIBP-03. SIBP-03 (0.01-10 μg/mL) specifically and concentration-dependently blocked both neuregulin (NRG)-dependent and -independent HER3 activation, attenuated HER3-mediated downstream signaling and inhibited cell proliferation. This antitumor activity was dependent, at least in part, on SIBP-03-induced, cell-mediated cytotoxicity and cellular phagocytosis. Importantly, SIBP-03 enhanced the antitumor activity of EGFR- or HER2-targeted drugs (cetuximab or trastuzumab) in vitro and in vivo. The mechanisms underlying this synergy involve increased inhibition of HER3-mediated downstream signaling. Collectively, these results demonstrated that SIBP-03, which is currently undergoing a Phase I clinical trial in China, may offer a new treatment option for patients with cancers harboring activated HER3, particularly as part of a combinational therapeutic strategy.
    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:三阴性乳腺癌(TNBC)是一个重要的临床挑战。化疗仍然是大部分TNBC患者的支柱,而耐药和肿瘤复发频繁发生。迫切需要确定TNBC的新分子靶标并开发针对侵袭性疾病的有效疗法。
    方法:进行免疫组织化学以检查TNBC样品中HER3的表达。蛋白质印迹用于评估蛋白质表达和活化。通过细胞生长(MTS)测定确定细胞增殖和活力。分析TCGA数据库以将HER3mRNA表达与TNBC患者的临床结果相关联。特异性shRNA用于敲低HER3表达。利用IncuCyte系统监测细胞生长和迁移。活/死细胞成像用于检测活细胞和死细胞。我们的抗HER3单克隆抗体(mAb)4A7的HER3识别通过ELISA验证,流式细胞术,和免疫共沉淀试验。在裸鼠中建立了原位肿瘤模型,以确定TNBC细胞形成肿瘤的能力,并测试我们的mAb4A7是否可以增强体内紫杉醇的抗肿瘤活性。
    结果:在测试的大约一半的TNBC样本和细胞系中观察到HER3的表达升高。对TCGA数据库的分析发现,在肿瘤中具有高HER3mRNA表达的TNBC患者显示出比具有低HER3表达的患者明显更差的总体生存率(OS)和无复发生存率(RFS)。HER3的特异性敲除在体外显著抑制TNBC细胞增殖和乳腺球形成以及在体内抑制肿瘤生长。我们的mAb4A7废除了heregulin(HER3的配体),但不是SDF-1(CXCR4的配体)诱导的TNBC细胞迁移的增强。4A7和EGFR酪氨酸激酶抑制剂(TKI)吉非替尼的组合显着降低磷酸化HER3,EGFR,Akt,和ERK1/2在TNBC细胞中并有效诱导生长抑制和细胞死亡。此外,4A7与紫杉醇的组合在体外和体内对TNBC具有显著的抗肿瘤活性。
    结论:我们的数据表明,增加的HER3是TNBC的有效治疗靶标,我们的抗HER3mAb(4A7)可能会增强吉非替尼或紫杉醇在TNBC中的疗效。
    BACKGROUND: Triple negative breast cancer (TNBC) represents a significant clinical challenge. Chemotherapy remains the mainstay for a large part of TNBC patients, whereas drug resistance and tumor recurrence frequently occur. It is in urgent need to identify novel molecular targets for TNBC and develop effective therapy against the aggressive disease.
    METHODS: Immunohistochemistry was performed to examine the expression of HER3 in TNBC samples. Western blots were used to assess protein expression and activation. Cell proliferation and viability were determined by cell growth (MTS) assays. TCGA databases were analyzed to correlate HER3 mRNA expression with the clinical outcomes of TNBC patients. Specific shRNA was used to knockdown HER3 expression. IncuCyte system was utilized to monitor cell growth and migration. LIVE/DEAD Cell Imaging was to detect live and dead cells. HER3 recognition by our anti-HER3 monoclonal antibody (mAb) 4A7 was verified by ELISA, flow cytometry, and co-immunoprecipitation assays. Orthotopic tumor models were established in nude mice to determine the capability of TNBC cells forming tumors and to test if our mAb 4A7 could potentiate the antitumor activity of paclitaxel in vivo.
    RESULTS: Elevated expression of HER3 was observed in approximately half of the TNBC specimens and cell lines tested. Analyses of TCGA databases found that the TNBC patients with high HER3 mRNA expression in the tumors showed significantly worse overall survival (OS) and relapse-free survival (RFS) than those with low HER3 expression. Specific knockdown of HER3 markedly inhibited TNBC cell proliferation and mammosphere formation in vitro and tumor growth in vivo. Our mAb 4A7 abrogated heregulin (a ligand for HER3), but not SDF-1 (a ligand for CXCR4)-induced enhancement of TNBC cell migration. Combinations of 4A7 and the EGFR-tyrosine kinase inhibitor (TKI) gefitinib dramatically decreased the levels of phosphorylated HER3, EGFR, Akt, and ERK1/2 in TNBC cells and potently induced growth inhibition and cell death. Moreover, 4A7 in combination with paclitaxel exerted significant antitumor activity against TNBC in vitro and in vivo.
    CONCLUSIONS: Our data demonstrate that increased HER3 is an effective therapeutic target for TNBC and our anti-HER3 mAb (4A7) may enhance the efficacy of gefitinib or paclitaxel in TNBC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    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
    BRAFV600E,最常见的基因改变,已成为甲状腺癌的主要治疗靶点。Vemurafenib(PLX4032),BRAFV600E激酶的特异性抑制剂,在BRAFV600E突变的甲状腺癌患者中表现出抗肿瘤活性。然而,PLX4032的临床获益通常受到短期缓解和通过异质性反馈机制获得的获得性耐药的限制.双硫仑(DSF),一种厌恶酒精的药物,以铜(Cu)依赖性方式显示有效的抗肿瘤功效。然而,其在甲状腺癌中的抗肿瘤活性及其对BRAF激酶抑制剂细胞反应的影响尚不清楚.通过一系列体外和体内功能实验,系统地评估了DSF/Cu对BRAFV600E突变的甲状腺癌细胞的抗肿瘤作用及其对这些细胞对BRAF激酶抑制剂PLX4032的反应的影响。通过Westernblot和流式细胞术检测,探讨了DSF/Cu对PLX4032致敏作用的分子机制。DSF/Cu对BRAFV600E突变的甲状腺癌细胞的增殖和集落形成的抑制作用强于单独的DSF处理。进一步的研究表明,DSF/Cu通过ROS依赖性抑制MAPK/ERK和PI3K/AKT信号通路杀死甲状腺癌细胞。我们的数据还表明,DSF/Cu显着增加了BRAFV600E突变的甲状腺癌细胞对PLX4032的反应。机械上,DSF/Cu通过以ROS依赖性方式抑制HER3和AKT并随后缓解MAPK/ERK和PI3K/AKT通路的反馈激活,使BRAF突变甲状腺癌细胞对PLX4032敏感。这项研究不仅暗示了DSF/Cu在癌症治疗中的潜在临床应用,而且为BRAFV600E突变的甲状腺癌提供了新的治疗策略。
    BRAFV600E, the most common genetic alteration, has become a major therapeutic target in thyroid cancer. Vemurafenib (PLX4032), a specific inhibitor of BRAFV600E kinase, exhibits antitumor activity in patients with BRAFV600E-mutated thyroid cancer. However, the clinical benefit of PLX4032 is often limited by short-term response and acquired resistance via heterogeneous feedback mechanisms. Disulfiram (DSF), an alcohol-aversion drug, shows potent antitumor efficacy in a copper (Cu)-dependent way. However, its antitumor activity in thyroid cancer and its effect on cellular response to BRAF kinase inhibitors remain unclear. Antitumor effects of DSF/Cu on BRAFV600E-mutated thyroid cancer cells and its effect on the response of these cells to BRAF kinase inhibitor PLX4032 were systematically assessed by a series of in vitro and in vivo functional experiments. The molecular mechanism underlying the sensitizing effect of DSF/Cu on PLX4032 was explored by Western blot and flow cytometry assays. DSF/Cu exhibited stronger inhibitory effects on the proliferation and colony formation of BRAFV600E-mutated thyroid cancer cells than DSF treatment alone. Further studies revealed that DSF/Cu killed thyroid cancer cells by ROS-dependent suppression of MAPK/ERK and PI3K/AKT signaling pathways. Our data also showed that DSF/Cu strikingly increased the response of BRAFV600E-mutated thyroid cancer cells to PLX4032. Mechanistically, DSF/Cu sensitizes BRAF-mutant thyroid cancer cells to PLX4032 by inhibiting HER3 and AKT in an ROS-dependent way and subsequently relieving feedback activation of MAPK/ERK and PI3K/AKT pathways. This study not only implies potential clinical use of DSF/Cu in cancer therapy but also provides a new therapeutic strategy for BRAFV600E-mutated thyroid cancers.
    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: Editorial
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号