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
    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
    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
    目的:寻找新的预测乳腺癌生物标志物。我们分析了参与致癌作用的生物标志物的血清浓度,也可以通过治疗靶向。
    方法:在单中心前瞻性研究中,AuroraA的血清水平,在新辅助治疗前,使用ELISA试剂盒测定了119名BC女性的胸苷激酶1和人表皮生长因子受体3型(HER3)。
    结果:分析以下临床资料:年龄;TNM;ER的表达,PGR,HER2和Ki67;组织学分级(G);以及在残余肿瘤负荷分类(RCB)中对新辅助治疗(NAT)的反应。NAT后41例患者(34%)获得了完整的病理反应(pCR)。确认pCR的患者比例最高的是三阴性乳腺癌(TNBC)(62.5%);非腔HER2阳性(52.6%)癌症亚型(p=0.0003);G3组(50%;p=0.0078)。AuroraA水平较高的患者更有可能达到pCR(p=0.039)。在多变量分析中,血清AuroraA水平≥4.75ng/mL与较高的pCR率相关(OR:3.5;95%CI:1.2-10.1;p=0.023).
    结论:我们表明,在一组生物异质性的BC患者中,治疗前血清AuroraA水平对预测NAT反应具有显著价值。
    OBJECTIVE: To search for new predictive breast cancer biomarkers. We analyzed the serum concentrations of biomarkers involved in carcinogenesis, which can also be targeted by therapy.
    METHODS: In a single-center prospective study, the serum levels of Aurora A, thymidine kinase 1, and human epidermal growth factor receptor type 3 (HER3) were determined in 119 women with BC before neoadjuvant treatment using ELISA kits.
    RESULTS: The following clinical data were analyzed: age; TNM; the expression of ER, PGR, HER2, and Ki67; histological grade (G); and the response to neoadjuvant treatment (NAT) in the residual tumor burden classification (RCB). A complete pathological response (pCR) was achieved after NAT in 41 patients (34%). The highest proportion of the patients with a confirmed pCR was found for triple negative breast cancer (TNBC) (62.5%); non-luminal HER2-positive (52.6%) cancer subtypes (p = 0.0003); and in the G3 group (50%; p = 0.0078). The patients with higher levels of Aurora A were more likely to achieve pCR (p = 0.039). In the multivariate analysis, the serum Aurora A levels ≥ 4.75 ng/mL correlated with a higher rate of pCR (OR: 3.5; 95% CI: 1.2-10.1; p = 0.023).
    CONCLUSIONS: We showed that in a biologically heterogeneous group of BC patients, the pretreatment serum Aurora A levels were of significant value in predicting the response to NAT.
    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:Patritumabderuxtecan(HER3-DXd)是一种针对人表皮生长因子受体3(HER3)的抗体-药物偶联物,由完全人抗HER3单克隆抗体(patritumab)通过稳定的负载共价连接到拓扑异构酶I抑制剂,肿瘤选择性,基于四肽的可裂解接头。TOT-HER3是一项旨在评估生物活性的机会之窗研究,通过CelTIL评分[=-0.8×肿瘤细胞性(%)+1.3×肿瘤浸润淋巴细胞(TIL)(%)]测量,在原发性可手术的HER2阴性早期乳腺癌患者中,短期(21天)术前治疗期间HER3-DXd的临床活性。
    方法:根据基线ERBB3信使RNA表达,将先前未经治疗的激素受体阳性/HER2阴性肿瘤的患者分为四个队列之一。所有患者均接受1剂HER3-DXd6.4mg/kg。主要目的是评估CelTIL评分从基线的变化。
    结果:评估了77例患者的疗效。观察到CelTIL评分的显著变化,中位数从基线增加3.5(四分位数间距,-3.8至12.7;P=0.003)。在可评估临床反应的患者中(n=62),观察到45%的总体反应率(通过卡尺测量肿瘤),与无响应者相比,响应者的CelTIL得分有增加的趋势(平均差,+11.9对+1.9)。CelTIL评分的变化与基线ERBB3信使RNA和HER3蛋白水平无关。基因组发生了变化,包括根据PAM50亚型转向增殖性较低的肿瘤表型,抑制细胞增殖基因,和诱导与免疫相关的基因。96%的患者(14%≥3级)观察到因治疗引起的不良事件;最常见的是恶心,疲劳,脱发,腹泻,呕吐,腹痛,中性粒细胞计数减少。
    结论:单剂量HER3-DXd与临床反应相关,增加的免疫浸润,抑制激素受体阳性/HER2阴性早期乳腺癌的增殖,和与以前报道的结果一致的耐受性安全性。这些发现支持HER3-DXd在早期乳腺癌中的进一步研究。
    Patritumab deruxtecan (HER3-DXd) is a human epidermal growth factor receptor 3 (HER3)-directed antibody-drug conjugate composed of a fully human anti-HER3 monoclonal antibody (patritumab) covalently linked to a topoisomerase I inhibitor payload via a stable, tumor-selective, tetrapeptide-based cleavable linker. TOT-HER3 is a window-of-opportunity study designed to assess the biological activity, measured by CelTIL score [= -0.8 × tumor cellularity (in %)  +  1.3  × tumor-infiltrating lymphocytes (TILs) (in %)], and clinical activity of HER3-DXd during short-term (21 days) pre-operative treatment in patients with primary operable HER2-negative early breast cancer.
    Patients with previously untreated hormone receptor-positive/HER2-negative tumors were allocated to one of four cohorts according to baseline ERBB3 messenger RNA expression. All patients received one dose of HER3-DXd 6.4 mg/kg. The primary objective was to evaluate change from baseline in CelTIL score.
    Seventy-seven patients were evaluated for efficacy. A significant change in CelTIL score was observed, with a median increase from baseline of 3.5 (interquartile range, -3.8 to 12.7; P = 0.003). Among patients assessable for clinical response (n = 62), an overall response rate of 45% was observed (tumor measurement by caliper), with a trend toward an increase in CelTIL score among responders compared with non-responders (mean difference, +11.9 versus +1.9). Change in CelTIL score was independent of baseline ERBB3 messenger RNA and HER3 protein levels. Genomic changes occurred, including switching toward a less proliferative tumor phenotype based on PAM50 subtypes, suppression of cell proliferation genes, and induction of genes associated with immunity. Treatment-emergent adverse events were observed in 96% of patients (14% grade ≥3); most common were nausea, fatigue, alopecia, diarrhea, vomiting, abdominal pain, and neutrophil count decrease.
    A single dose of HER3-DXd was associated with clinical response, increased immune infiltration, suppression of proliferation in hormone receptor-positive/HER2-negative early breast cancer, and a tolerable safety profile consistent with previously reported results. These findings support further study of HER3-DXd in early 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: Clinical Trial
    Sym013含有六种人源化单克隆抗体,其结合三种人表皮生长因子受体(HER1-3)上的非重叠表位。临床前研究表明Sym013强烈抑制多发性上皮肿瘤的生长。这是一项首次在人类研究中探索Sym013在晚期上皮恶性肿瘤患者中的安全性和有效性。
    剂量递增使用单患者队列,直到满足停止规则,其次是3+3设计。计划的剂量水平为:1、2、4、6、9、12、15和18mg/kg。治疗周期为28天,每八周成像。在多个时间点收集血清样品以评估药代动力学和抗药物抗体的开发。
    32例患者纳入多发性实体瘤,最常见的是结直肠癌(CRC;10/32,31%)。由于粘膜炎,皮疹,和腹泻4毫克/公斤,每周一次,剂量改为每两周一次(Q2W)。由于3级输注相关反应和9mg/kgQ2W的口腔粘膜炎,增加了强制性预防。当研究因商业原因终止时,15mg/kgQ2W队列正在注册。最常见的不良事件是皮肤(81%)和胃肠道(75%)疾病,包括皮炎/皮疹,口腔炎,和腹泻。1例CRC患者获得部分缓解;12例恶性肿瘤患者病情稳定。
    在研究进行期间,频繁输液相关反应的管理,皮肤毒性,粘膜疾病,这表明了她的抑制作用,需要多个协议修正案。调查人员,与赞助商合作,一致认为不可能实现具有可接受的目标饱和度的耐受方案.
    www.
    gov;NCT02906670(2016年9月20日)。
    Sym013 contains six humanized monoclonal antibodies that bind to non-overlapping epitopes on three human epidermal growth factor receptors (HER1-3). Preclinical studies suggested Sym013 strongly suppresses growth of multiple epithelial tumors. This is a first-in-human study exploring safety and efficacy of Sym013 in patients with advanced epithelial malignancies.
    Dose escalation used single-patient cohorts until the stopping rule was met, followed by 3 + 3 design. Dose levels planned were: 1, 2, 4, 6, 9, 12, 15, and 18 mg/kg. Treatment cycles were 28 days with imaging every eight weeks. Serum samples were collected at multiple time points for assessment of pharmacokinetics and development of anti-drug antibodies.
    Thirty-two patients were enrolled with multiple solid tumors, most common being colorectal cancer (CRC; 10/32, 31%). Due to mucositis, rash, and diarrhea at 4 mg/kg once-weekly, dosing was changed to biweekly (Q2W). Mandatory prophylaxis was added due to Grade 3 infusion-related reaction and oral mucositis at 9 mg/kg Q2W. The 15 mg/kg Q2W cohort was enrolling when the study was terminated for business reasons. Most common adverse events were skin (81%) and gastrointestinal (75%) disorders, including dermatitis/rash, stomatitis, and diarrhea. One patient with CRC achieved a partial response; 12 patients with varied malignancies had stable disease.
    During the conduct of the study, management of frequent infusion-related reactions, skin toxicities, and mucosal disorders, which are indicative of HER inhibition, necessitated multiple protocol amendments. The investigators, in concert with the Sponsor, agreed that achieving a tolerated regimen with acceptable target saturation was unlikely.
    www.
    gov ; NCT02906670 (September 20, 2016).
    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
    Background Overactivation of human epidermal growth factor receptor 3 (HER3) triggers multiple intracellular pathways resulting in tumor cell survival. This Phase 1 study assessed the safety, efficacy, and pharmacokinetics (PK) of seribantumab, a fully human anti-HER3 monoclonal antibody. Methods Adult patients with advanced or refractory solid tumors were treated in six dose cohorts of seribantumab: 3.2, 6, 10, 15, or 20 mg/kg weekly, or 40 mg/kg loading dose followed by 20 mg/kg weekly maintenance dose (40/20 mg/kg) using a modified 3 + 3 dose escalation strategy with cohort expansion. Primary objectives were identification of a recommended Phase 2 dose (RP2D) and determination of objective response rate. Secondary objectives were assessment of safety, dose-limiting toxicities, and PK. Results Forty-four patients (26 dose escalation; 18 dose expansion) were enrolled. Seribantumab monotherapy was well tolerated with most adverse events being transient and mild to moderate (grade 1 or 2) in severity; maximum tolerated dose was not reached. The highest dose, 40/20 mg/kg, was identified as RP2D. Best response was stable disease, reported in 24% and 39% of patients during the dose escalation and expansion portions of the study, respectively. Seribantumab terminal half-life was ≈100 h; steady state concentrations were reached after 3-4 weekly doses. Conclusions Seribantumab monotherapy was well tolerated across all dose levels. Safety and PK data from this study support further seribantumab investigations in genomically defined populations.Clinical trial registration NCT00734305. August 12, 2008.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Clinical Trial, Phase I
    GSK2849330,一种抗HER3单克隆抗体,可阻断癌细胞中的HER3/Neuregulin1(NRG1)信号传导,被设计用于增强的抗体依赖性细胞毒性和补体依赖性细胞毒性。这个阶段I,人类第一,开放标签研究评估了安全性,药代动力学(PK),药效学,GSK2849330在表达HER3的晚期实体瘤患者中的初步活性。
    通过免疫组织化学前瞻性选择具有各种肿瘤类型的患者的HER3表达;还筛选了NRG1mRNA表达的子集。在剂量递增阶段,患者每2周接受GSK28493301.4-30mg/kg,或每周3mg/kg或30mg/kg,静脉注射(IV)。在剂量膨胀阶段,患者每周静脉注射30mg/kgGSK2849330.
    29例HER3表达癌症患者,其中2人表达NRG1,接受GSK2849330(剂量递增:n=18,剂量扩大:n=11)。GSK2849330耐受性良好。没有观察到剂量限制性毒性。最高剂量,每周30毫克/千克,预计将提供全面的目标参与,被选择用于剂量扩张。治疗引起的不良事件(AE)大多为1级或2级。最常见的不良事件是腹泻(66%),疲劳(62%),食欲下降(31%)。实现了与剂量成正比的血浆暴露,在配对的组织活检中有HER3抑制的证据。29名患者中,只有1个确认部分反应,持续19个月,在CD74-NRG1重排的非小细胞肺癌(NSCLC)患者中发现。
    GSK2849330表现出良好的安全性,剂量比例PK,和目标交战的证据,但在表达HER3的癌症中抗肿瘤活性有限。在CD74-NRG1重排的NSCLC患者中观察到的异常反应表明在NRG1融合阳性癌症中的进一步探索。
    这项首次人体研究证实GSK2849330具有良好的耐受性。重要的是,在各种表达HER3的晚期肿瘤中,仅通过HER3/NRG1表达进行前瞻性选择不足以鉴定可从这种抗体依赖性细胞介导的细胞毒性和补体依赖性细胞毒性增强的抗HER3抗体治疗中获益的患者。唯一确认的持久反应是CD74-NRG1重排肺癌患者。这凸显了在正在进行的试验中,前瞻性地跨肿瘤类型筛选NRG1融合体以丰富抗HER3药物的潜在应答者的潜在效用。
    GSK2849330, an anti-HER3 monoclonal antibody that blocks HER3/Neuregulin 1 (NRG1) signaling in cancer cells, is engineered for enhanced antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity. This phase I, first-in-human, open-label study assessed the safety, pharmacokinetics (PK), pharmacodynamics, and preliminary activity of GSK2849330 in patients with HER3-expressing advanced solid tumors.
    Patients with various tumor types were prospectively selected for HER3 expression by immunohistochemistry; a subset was also screened for NRG1 mRNA expression. In the dose-escalation phase, patients received GSK2849330 1.4-30 mg/kg every 2 weeks, or 3 mg/kg or 30 mg/kg weekly, intravenously (IV). In the dose-expansion phase, patients received 30 mg/kg GSK2849330 IV weekly.
    Twenty-nine patients with HER3-expressing cancers, of whom two expressed NRG1, received GSK2849330 (dose escalation: n = 18, dose expansion: n = 11). GSK2849330 was well tolerated. No dose-limiting toxicities were observed. The highest dose, of 30 mg/kg weekly, expected to provide full target engagement, was selected for dose expansion. Treatment-emergent adverse events (AEs) were mostly grade 1 or 2. The most common AEs were diarrhea (66%), fatigue (62%), and decreased appetite (31%). Dose-proportional plasma exposures were achieved, with evidence of HER3 inhibition in paired tissue biopsies. Of 29 patients, only 1 confirmed partial response, lasting 19 months, was noted in a patient with CD74-NRG1-rearranged non-small cell lung cancer (NSCLC).
    GSK2849330 demonstrated a favorable safety profile, dose-proportional PK, and evidence of target engagement, but limited antitumor activity in HER3-expressing cancers. The exceptional response seen in a patient with CD74-NRG1-rearranged NSCLC suggests further exploration in NRG1-fusion-positive cancers.
    This first-in-human study confirms that GSK2849330 is well tolerated. Importantly, across a variety of HER3-expressing advanced tumors, prospective selection by HER3/NRG1 expression alone was insufficient to identify patients who could benefit from treatment with this antibody-dependent cell-mediated cytotoxicity- and complement-dependent cytotoxicity-enhanced anti-HER3 antibody. The only confirmed durable response achieved was in a patient with CD74-NRG1-rearranged lung cancer. This highlights the potential utility of screening for NRG1 fusions prospectively across tumor types to enrich potential responders to anti-HER3 agents in ongoing trials.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Background: Preclinical data support a key role for the human epidermal growth factor receptor 3 (HER3) pathway in hormone receptor (HR)-positive breast cancer. Recently, new HER3 directed antibody drug conjugates have shown activity in breast cancer. Given the need to better understand the molecular biology, tumor microenvironment, and mechanisms of drug resistance in breast cancer, we designed this window-of-opportunity study with the HER3 directed antibody drug conjugate patritumab deruxtecan (HER3-DXd; U3-1402). Trial Design: Based on these data, a prospective, multicenter, single-arm, window-of-opportunity study was designed to evaluate the biological effect of patritumab deruxtecan in the treatment of naïve patients with HR-positive/HER2-negative early breast cancer whose primary tumors are ≥1 cm by ultrasound evaluation. Patients will be enrolled in four cohorts according to the mRNA-based ERBB3 expression by central assessment. The primary endpoint is a CelTIL score after one single dose. A translational research plan is also included to provide biological information and to evaluate secondary and exploratory objectives of the study. Trial Registration Number: EudraCT 2019-004964-23; NCT number: NCT04610528.
    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
    尽管有催化缺陷,假激酶通常是细胞信号传导的重要参与者,充当其活性对应物的变构调节剂。越来越多的假激酶的失调与人类疾病有关,制造感兴趣的假激酶治疗靶标。假激酶可以是动态的,采用对其变构功能至关重要的特定构象。干扰它们的变构作用,小分子会锁定假激酶的构象,阻止它们的生产性伴侣相互作用,是一种有吸引力的治疗策略。作为众所周知的表皮生长因子受体家族成员的变构激活剂,在癌症进展中起着重要作用,假激酶HER3是解决假激酶作为变构抑制剂开发的药物靶标的潜力的相关背景。在这个概念验证研究中,我们开发了一个多元,中等通量热转移测定筛选策略,以评估超过100000种化合物,并鉴定选择性小分子抑制剂,这些抑制剂会将HER3捕获在不利于活性HER2-HER3异二聚体形成的构象中。作为一种概念验证化合物,AC3573以一定的特异性与HER3结合并消除细胞中的HER2-HER3复合物形成和下游信号传导。我们的研究强调了确定干扰假激酶生物学功能的新分子作用机制的机会。
    Despite being catalytically defective, pseudokinases are typically essential players of cellular signalling, acting as allosteric regulators of their active counterparts. Deregulation of a growing number of pseudokinases has been linked to human diseases, making pseudokinases therapeutic targets of interest. Pseudokinases can be dynamic, adopting specific conformations critical for their allosteric function. Interfering with their allosteric role, with small molecules that would lock pseudokinases in a conformation preventing their productive partner interactions, is an attractive therapeutic strategy to explore. As a well-known allosteric activator of epidermal growth factor receptor family members, and playing a major part in cancer progression, the pseudokinase HER3 is a relevant context in which to address the potential of pseudokinases as drug targets for the development of allosteric inhibitors. In this proof-of-concept study, we developed a multiplex, medium-throughput thermal shift assay screening strategy to assess over 100 000 compounds and identify selective small molecule inhibitors that would trap HER3 in a conformation which is unfavourable for the formation of an active HER2-HER3 heterodimer. As a proof-of-concept compound, AC3573 bound with some specificity to HER3 and abrogated HER2-HER3 complex formation and downstream signalling in cells. Our study highlights the opportunity to identify new molecular mechanisms of action interfering with the biological function of pseudokinases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号