HER3

HER3
  • 文章类型: Journal Article
    目的:HER3,EGFR受体家族成员,在驱动乳腺癌的致癌细胞增殖中起着核心作用。新型HER3疗法显示出有希望的结果,而最近开发的HER3PET成像方式有助于预测和评估早期治疗反应。然而,基线HER3表达,以及新辅助治疗时表达的变化,没有得到很好的表征。我们进行了一项前瞻性临床研究,新辅助/全身治疗前后,在新诊断的乳腺癌患者中确定HER3表达,并确定通过HER3受体维持的可能的抗性机制。
    方法:该研究于2018年5月25日至2019年10月12日进行。34例新诊断的任何亚型乳腺癌患者(ER±,PR±,HER2±)纳入研究。在诊断时从每个患者获得两个核心活检标本。四名患者在开始新辅助/全身治疗或全身治疗后接受了第二次研究活检,我们将其定义为新辅助治疗。在治疗开始之前和之后进行HER3和PI3K/AKT和MAPK途径的下游信号传导节点的分子表征。在外部数据集(GSE122630)中进行筛选的转录验证。
    结果:在新诊断的乳腺癌中发现了不同的基线HER3表达,并且与pAKT呈正相关(r=0.45)。在接受新辅助/全身治疗的患者中,HER3表达的变化是可变的。在激素受体阳性(ER/PR/HER2-)患者中,新辅助治疗后HER3表达有统计学意义的增加,而ER+/PR+/HER2+患者的HER3表达无显著变化。然而,这两名患者均显示PI3K/AKT通路下游信号传导增加.一名患有ER+/PR-/HER2-乳腺癌的受试者和另一名患有ER+/PR+/HER2+乳腺癌的受试者显示出降低的HER3表达。转录组发现,显示治疗后HER3表达降低的患者存在免疫抑制环境。
    结论:本研究证实了不同乳腺癌亚型的HER3表达。HER3表达可以早期评估,新辅助治疗后,为癌症生物学提供有价值的见解,并可能作为预后生物标志物。新辅助治疗评估的临床转化可以使用HER3PET成像来实现,提供有关肿瘤生物学的实时信息,并指导乳腺癌患者的个性化治疗。
    OBJECTIVE: HER3, a member of the EGFR receptor family, plays a central role in driving oncogenic cell proliferation in breast cancer. Novel HER3 therapeutics are showing promising results while recently developed HER3 PET imaging modalities aid in predicting and assessing early treatment response. However, baseline HER3 expression, as well as changes in expression while on neoadjuvant therapy, have not been well-characterized. We conducted a prospective clinical study, pre- and post-neoadjuvant/systemic therapy, in patients with newly diagnosed breast cancer to determine HER3 expression, and to identify possible resistance mechanisms maintained through the HER3 receptor.
    METHODS: The study was conducted between May 25, 2018 and October 12, 2019. Thirty-four patients with newly diagnosed breast cancer of any subtype (ER ± , PR ± , HER2 ±) were enrolled in the study. Two core biopsy specimens were obtained from each patient at the time of diagnosis. Four patients underwent a second research biopsy following initiation of neoadjuvant/systemic therapy or systemic therapy which we define as neoadjuvant therapy. Molecular characterization of HER3 and downstream signaling nodes of the PI3K/AKT and MAPK pathways pre- and post-initiation of therapy was performed. Transcriptional validation of finings was performed in an external dataset (GSE122630).
    RESULTS: Variable baseline HER3 expression was found in newly diagnosed breast cancer and correlated positively with pAKT across subtypes (r = 0.45). In patients receiving neoadjuvant/systemic therapy, changes in HER3 expression were variable. In a hormone receptor-positive (ER +/PR +/HER2-) patient, there was a statistically significant increase in HER3 expression post neoadjuvant therapy, while there was no significant change in HER3 expression in a ER +/PR +/HER2+ patient. However, both of these patients showed increased downstream signaling in the PI3K/AKT pathway. One subject with ER +/PR -/HER2- breast cancer and another subject with ER +/PR +/HER2 + breast cancer showed decreased HER3 expression. Transcriptomic findings, revealed an immune suppressive environment in patients with decreased HER3 expression post therapy.
    CONCLUSIONS: This study demonstrates variable HER3 expression across breast cancer subtypes. HER3 expression can be assessed early, post-neoadjuvant therapy, providing valuable insight into cancer biology and potentially serving as a prognostic biomarker. Clinical translation of neoadjuvant therapy assessment can be achieved using HER3 PET imaging, offering real-time information on tumor biology and guiding personalized treatment for breast cancer patients.
    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
    乳腺癌是一种异质性疾病,包含多个不同的亚型。由于对每个亚型的不同生物学特征的了解不断增加,大多数患者接受基于已知生物标志物的个性化治疗.然而,一些生物标志物在乳腺癌进化中的作用仍然未知,它们作为治疗靶点的潜在用途仍未得到充分开发。HER3是人类表皮生长因子受体家族的一员,在50%-70%的乳腺癌中过表达。HER3在癌症进展中起关键作用,转移发展,以及所有乳腺癌亚型的耐药性。由于其在癌症进展中的关键作用,在过去的10年中,许多HER3靶向疗法的研发结果相互矛盾.下一代抗体-药物缀合物最近在表达HER3的实体瘤(包括乳腺癌)中显示有希望的结果。在这次审查中,我们讨论了HER3在乳腺癌发病机制中的作用及其在所有亚型中的相关性.我们还探索了新的抗HER3治疗策略,质疑HER3检测作为乳腺癌治疗中重要信息的重要性。
    Breast cancer is a heterogeneous disease, encompassing multiple different subtypes. Thanks to the increasing knowledge of the diverse biological features of each subtype, most patients receive personalized treatment based on known biomarkers. However, the role of some biomarkers in breast cancer evolution is still unknown, and their potential use as a therapeutic target is still underexplored. HER3 is a member of the human epidermal growth factors receptor family, overexpressed in 50%-70% of breast cancers. HER3 plays a key role in cancer progression, metastasis development, and drug resistance across all the breast cancer subtypes. Owing to its critical role in cancer progression, many HER3-targeting therapies have been developed over the past decade with conflicting findings. Next-generation antibody-drug conjugates have recently shown promising results in solid tumors expressing HER3, including breast cancer. In this review, we discuss the HER3 role in the pathogenesis of breast cancer and its relevance across all subtypes. We also explore the new anti-HER3 treatment strategies, calling into question the significance of HER3 detection as crucial information in breast cancer treatment.
    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
    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:脂肪肉瘤是最常见的间充质恶性肿瘤之一。然而,治疗选择仍然非常有限,到目前为止,靶向治疗尚未建立.免疫疗法,这是其他肿瘤实体的突破,似乎对脂肪肉瘤没有疗效。使事情进一步复杂化,分类仍然是困难的,由于形态学的多样性和非特异性或缺乏在免疫组织化学标记,使用FISH或测序作为最佳选择的分子病理学。许多脂肪肉瘤携带MDM2基因扩增。与MDM2的基因座密切相关,存在HER3(ERBB3)基因,并且可以发生共扩增。由于HER/EGFR受体酪氨酸激酶及其抑制剂/抗体在广泛的肿瘤疾病和治疗中起作用,一些HER3抑制剂/抗体已经在临床研究中,我们假设在HER3共扩增的情况下,肿瘤可能具有进一步的潜在治疗靶点.
    方法:我们对56例存档病例进行了FISH分析(MDM2、DDIT3、HER3),随后进行了重新分类以确认脂肪肉瘤的诊断。
    结果:在56例病例中,有16例需要重新分类,在54个案例中,有20个,可以检测到HER3的簇扩增,与MDM2扩增显着相关。我们的研究表明,脂肪肉瘤的实体显示出特定的分子特征,导致现代,既定的方法论。此外,在57.1%的病例中,HER3被大量簇扩增,为靶向治疗提供了一个推定的治疗靶点。
    结论:我们的研究为进一步研究HER3基因作为脂肪肉瘤的假定治疗靶点奠定了基础。
    BACKGROUND: Liposarcomas are among the most common mesenchymal malignancies. However, the therapeutic options are still very limited and so far, targeted therapies had not yet been established. Immunotherapy, which has been a breakthrough in other oncological entities, seems to have no efficacy in liposarcoma. Complicating matters further, classification remains difficult due to the diversity of morphologies and nonspecific or absent markers in immunohistochemistry, leaving molecular pathology using FISH or sequencing as best options. Many liposarcomas harbor MDM2 gene amplifications. In close relation to the gene locus of MDM2, HER3 (ERBB3) gene is present and co-amplification could occur. Since the group of HER/EGFR receptor tyrosine kinases and its inhibitors/antibodies play a role in a broad spectrum of oncological diseases and treatments, and some HER3 inhibitors/antibodies are already under clinical investigation, we hypothesized that in case of HER3 co-amplifications a tumor might bear a further potential therapeutic target.
    METHODS: We performed FISH analysis (MDM2, DDIT3, HER3) in 56 archived cases and subsequently performed reclassification to confirm the diagnosis of liposarcoma.
    RESULTS: Next to 16 out of 56 cases needed to be re-classified, in 20 out of 54 cases, a cluster-amplification of HER3 could be detected, significantly correlating with MDM2 amplification. Our study shows that the entity of liposarcomas show specific molecular characteristics leading to reclassify archived cases by modern, established methodologies. Additionally, in 57.1% of these cases, HER3 was cluster-amplified profusely, presenting a putative therapeutic target for targeted therapy.
    CONCLUSIONS: Our study serves as the initial basis for further investigation of the HER3 gene as a putative therapeutic target in liposarcoma.
    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
    取决于癌症类型,HER3在~2%-10%的癌症中突变。我们发现HER3-V104L突变从HER3KOHER2+HCC1569乳腺癌细胞中通过慢病毒转导引入的患者衍生突变激活,其中内源性HER3被CRISPR/Cas9消除。与表达野生型HER3或HER3-V104M的细胞相比,表达HER3-V104L的细胞显示更高的p-HER3和p-ERK1/2表达。与缺乏HER3突变的患者相比,肿瘤表达HER3V104L变体的患者的总体生存率降低,而我们没有发现具有HER3V104M突变的各种癌症患者的总体生存率有统计学上的显着差异。我们的数据显示HER2抑制剂抑制稳定表达HER3-V104L突变的HCC1569HER3KO细胞的细胞生长。具有内源性HER3-V104M突变的癌细胞系(SNU407、UC15和DV90)显示用HER2抑制剂处理的细胞增殖和p-HER2/p-ERK1/2表达降低。与PI3K抑制剂相比,敲低HER3消除了对ERK抑制剂SCH779284总体更敏感的上述细胞系中的细胞增殖。HER3-V104L突变稳定了COS7和SNUC5细胞中HER3蛋白的表达。在HER结合配偶体存在下瞬时转染HER3-V104L突变的COS7细胞以不依赖NRG的方式显示p-HER3、p-ERK1/2相对于HER3-WT的更高表达,而AKT信号传导没有任何变化。总的来说,本研究显示了HER3V104L和V104M突变的临床意义及其对HER2,PI3K和ERK抑制剂的反应.
    HER3 is mutated in ~2%-10% of cancers depending on the cancer type. We found the HER3-V104L mutation to be activating from patient-derived mutations introduced via lentiviral transduction in HER3KO HER2 + HCC1569 breast cancer cells in which endogenous HER3 was eliminated by CRISPR/Cas9. Cells expressing HER3-V104L showed higher p-HER3 and p-ERK1/2 expression versus cells expressing wild-type HER3 or HER3-V104M. Patients whose tumor expressed the HER3 V104L variant had a reduced probability of overall survival compared to patients lacking a HER3 mutation whereas we did not find a statistically significant difference in overall survival of various cancer patients with the HER3 V104M mutation. Our data showed that HER2 inhibitors suppressed cell growth of HCC1569HER3KO cells stably expressing the HER3-V104L mutation. Cancer cell lines (SNU407, UC15 and DV90) with endogenous HER3-V104M mutation showed reduced cell proliferation and p-HER2/p-ERK1/2 expression with HER2 inhibitor treatment. Knock down of HER3 abrogated cell proliferation in the above cell lines which were overall more sensitive to the ERK inhibitor SCH779284 versus PI3K inhibitors. HER3-V104L mutation stabilized HER3 protein expression in COS7 and SNUC5 cells. COS7 cells transiently transfected with the HER3-V104L mutation in the presence of HER binding partners showed higher expression of p-HER3, p-ERK1/2 versus HER3-WT in a NRG-independent manner without any change in AKT signaling. Overall, this study shows the clinical relevance of the HER3 V104L and the V104M mutations and its response to HER2, PI3K and ERK 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
    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
    间变性淋巴瘤激酶(ALK)靶向酪氨酸激酶抑制剂(TKIs)可改善患者生存率;然而,一些患者出现ALK-TKI耐药的机制不明.我们调查了ALK阳性非小细胞肺癌(NSCLC)患者的ErbB家族和c-MET表达,以了解它们在ALK-TKI反应中的作用。
    我们研究了72例晚期ALK阳性非小细胞肺癌患者的EML4-ALK融合变异亚型和c-MET免疫染色,EGFR,组织标本上的HER2和HER3在用ALK-TKI治疗前(初次)和后(二次)。我们研究了它们的表达与生存结果的关系,并评估了ALK和EGFR抑制剂在HER3特异性配体HRG1刺激的ALK阳性NSCLC细胞系中的有效性。
    c-MET的高表达,EGFR,HER2和HER3在4.9%中观察到,18.0%,1.6%,和25.8%的原发性肿瘤,分别,和18.5%,37.0%,10.7%,和35.7%的继发性肿瘤,分别。原发性肿瘤中的HER3过表达显示较差的生存率(P=0.132)。在具有EML4-ALK变体1/2(V1/V2)的亚组中,HER3过表达与原发性和继发性肿瘤的低生存率显著相关(分别为P=0.022和P=0.004)。氯拉替尼和厄洛替尼的联合治疗显著降低了ALK阳性NSCLC细胞中HRG1诱导的RTK信号激活。
    HER3过表达可能作为ALK阳性NSCLC的预后标志物,包括ALK-TKI幼稚和治疗病例,特别是那些与EML4-ALKV1/V2。评估HER3表达对于这些患者的治疗计划和结果预测可能至关重要。
    UNASSIGNED: Anaplastic lymphoma kinase (ALK)-targeted tyrosine kinase inhibitors (TKIs) improve patient survival; however, some patients develop ALK-TKI resistance with unidentified mechanisms. We investigated ErbB family and c-MET expression in patients with ALK-positive non-small cell lung cancer (NSCLC) to understand their roles in the ALK-TKI response.
    UNASSIGNED: We studied 72 patients with advanced ALK-positive NSCLC with EML4-ALK fusion variant subtyping and immunostaining for c-MET, EGFR, HER2, and HER3 on tissue specimens both pre- (primary) and post-treatment (secondary) with ALK-TKI. We investigated the association of their expression with survival outcomes and assessed the effectiveness of combining ALK and EGFR inhibitors in ALK-positive NSCLC cell lines stimulated with the HER3-specific ligand HRG1.
    UNASSIGNED: High expression of c-MET, EGFR, HER2, and HER3 was observed in 4.9%, 18.0%, 1.6%, and 25.8% of primary tumors, respectively, and 18.5%, 37.0%, 10.7%, and 35.7% of secondary tumors, respectively. HER3 overexpression in primary tumors showed inferior survival (P=0.132). In the subgroup with EML4-ALK variant 1/2 (V1/V2), HER3 overexpression was significantly associated with inferior survival in both primary and secondary tumors (P=0.022 and P=0.004, respectively). Combination treatment with lorlatinib and erlotinib significantly reduced HRG1-induced activation of RTK signaling in ALK-positive NSCLC cells.
    UNASSIGNED: HER3 overexpression has potential as a prognostic marker in ALK-positive NSCLCs, including ALK-TKI naïve and treated cases, especially those with EML4-ALK V1/V2. Assessing HER3 expression may be crucial for treatment planning and outcome prediction in these patients.
    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
    神经调节素1(NRG1)融合是在非小细胞肺癌(NSCLC)中检测到的致癌驱动因素,胰腺导管腺癌(PDAC)和其他实体瘤。NRG1融合是罕见的,发生在不到1%的实体瘤中。NRG1融合阳性(NRG1)癌症患者的治疗选择有限。Zenocutuzumab是一种新颖的,双特异性IgG1抗体,靶向HER2和HER3蛋白并通过“Dock&Block®”作用机制抑制NRG1结合。这里,我们描述了eNRGy试验第二阶段组成部分的基本原理和设计,整体的一部分,开放标签阶段I/II,探索安全性的多中心试验,耐受性,药代动力学,药效学,zenocutuzumab在NRG1+NSCLC患者中的免疫原性和抗肿瘤活性,PDAC或其他实体瘤。
    NRG1基因融合是导致癌细胞生长的罕见突变。这些融合在许多不同类型的癌症中发现。具有NRG1基因融合的肿瘤对标准治疗方案的反应不佳。Zenocutuzumab,或者芝诺,是一种正在测试的治疗方法,看看它是否可以阻止由于NRG1基因融合而生长的癌症。这里,我们描述了一项正在进行的临床试验(eNRGy)的推理和设计,该试验旨在研究Zeno在NRG1基因融合癌症患者中的疗效(效果)和安全性.eNRGy试验正在招募具有NRG1基因融合的癌症患者,包括非小细胞肺癌,胰腺癌和其他。参加这项试验的患者将每两周接受一次Zeno,直到他们的癌症生长。该试验的主要目标(主要终点)是确定肿瘤大小减少30%或更多的患者的百分比。eNRGy试验目前正在招募患者。有关更多信息,参考ClinicalTrials.gov(标识符:NCT02912949),访问https://nrg1.com/,或致电1-833-NRG-1234。
    Neuregulin 1 (NRG1) fusions are oncogenic drivers that have been detected in non-small-cell lung cancer (NSCLC), pancreatic ductal adenocarcinoma (PDAC) and other solid tumors. NRG1 fusions are rare, occurring in less than 1% of solid tumors. Patients with NRG1 fusion positive (NRG1+) cancer have limited therapeutic options. Zenocutuzumab is a novel, bispecific IgG1 antibody that targets both HER2 and HER3 proteins and inhibits NRG1 binding through a \'Dock & Block®\' mechanism of action. Here, we describe the rationale and design of the phase II component of the eNRGy trial, part of the overall, open-label phase I/II, multicenter trial exploring the safety, tolerability, pharmacokinetics, pharmacodynamics, immunogenicity and antitumor activity of zenocutuzumab in patients with NRG1+ NSCLC, PDAC or other solid tumors.
    eNRGy: a clinical trial of zenocutuzumab for cancer caused by NRG1 gene fusionsNRG1 gene fusions are rare mutations that cause cancer cells to grow. These fusions are found in many different types of cancer. Tumors with NRG1 gene fusions do not respond well to standard treatment options. Zenocutuzumab, or Zeno, is a treatment that is being tested to see if it can stop cancer that is growing because of NRG1 gene fusions. Here, we describe the reasoning for and design of an ongoing clinical trial (eNRGy) designed to study the efficacy (how well it works) and safety of Zeno in patients with cancer that has NRG1 gene fusions. The eNRGy trial is recruiting patients with cancer that has NRG1 gene fusions, including non-small-cell lung cancer, pancreatic cancer and others. Patients who join this trial will receive Zeno once every 2 weeks until their cancer grows. The main goal (primary end point) of this trial is to determine the percentage of patients whose tumors decrease in size by 30% or more. The eNRGy trial is currently enrolling patients. For more information, refer to ClinicalTrials.gov (Identifier: NCT02912949), visit https://nrg1.com/, or call 1-833-NRG-1234.
    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
    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

公众号