trastuzumab

曲妥珠单抗
  • 文章类型: Journal Article
    辅助化疗引起的心脏毒性(CTX)是影响乳腺癌(BC)患者预后和生活质量的重要因素。在这项研究中,我们旨在研究接受蒽环类药物辅助化疗方案(AC-T)和蒽环类药物联合曲妥珠单抗辅助化疗方案(AC-TH)的BC患者治疗前后心外膜脂肪组织(EAT)的变化.此外,我们评估了两组患者的EAT变化是否存在差异.我们的目的是检查蒽环类和曲妥珠单抗对EAT的影响,并确定EAT变化对CTX的潜在作用。
    我们回顾了接受AC-T和AC-TH辅助化疗方案治疗的女性BC患者,所有患者均接受基线(T0)和随访(T1)胸部计算机断层扫描(CT)和超声心动图检查.一群健康的女性,年龄匹配,做了两次胸部CT.使用半自动软件在胸部CT上定量EAT。CTX定义为左心室射血分数(LVEF)从基线下降>10%,绝对值<53%。
    本研究共纳入41例BC患者,AC-T组23例,AC-TH组18例。此外,22例健康女性作为正常组。BC患者均未在化疗后发生CTX。正常组与AC-T组(p=0.341)或AC-TH组(p=0.853)之间的年龄没有显着差异。同样,化疗前后,正常组的体重指数(BMI)与AC-T组(p=0.377,0.346)和AC-TH组(p=0.148,0.119)相当。AC-T组的EAT体积指数(mL/kg/m2)均显着较高(5.11±1.85vs.4.34±1.55,p<0.001)和AC-TH组(4.53±1.61vs.与T0相比,T1时3.48±1.62,p<0.001)。此外,AC-T组(-72.95±5.01vs.-71.22±3.91,p=0.005)和AC-TH组(-72.55±5.27vs.-68.20±5.98,p<0.001)与T0相比,T1时的EAT放射密度(HU)显着降低。然而,正常组无显著差异。在T0时,EAT体积指数没有差异(4.34±1.55与3.48±1.62,p=0.090)和放射性密度(-71.22±3.91vs.-68.20±5.98,p=0.059)在AC-T和AC-TH组之间。同样,在T1时,EAT体积指数(-5.11±1.85vs.4.53±1.61,p=0.308)和放射性密度(-72.95±5.00vs.-72.54±5.27,p=0.802)。
    接受AC-T和AC-TH辅助化疗方案的BC患者显示EAT体积指数显著上升,随着化疗后放射性密度的大幅降低。这些发现表明,EAT的改变可能有助于识别由化学治疗剂引起的心脏并发症,并提醒临床医生关注BC患者辅助化疗后EAT的变化,以防止CTX的实际发生。
    UNASSIGNED: Cardiotoxicity (CTX) induced by adjuvant chemotherapy is a significant factor that impacts the prognosis and quality of life in breast cancer (BC) patients. In this study, we aimed to investigate the changes in epicardial adipose tissue (EAT) before and after treatment in BC patients who received anthracyclines adjuvant chemotherapy protocol (AC-T) and anthracyclines combined with trastuzumabadjuvant chemotherapy protocol (AC-TH). Additionally, we assessed whether there were any differences in the changes in EAT between the two groups of patients. Our objective was to examine the effects of anthracyclines and trastuzumab on EAT and determine the potential role of EAT changes on CTX.
    UNASSIGNED: We reviewed female BC patients who were treated with adjuvant chemotherapy protocols of AC-T and AC-TH, all of whom underwent baseline (T0) and follow-up (T1) chest computed tomography (CT) and echocardiography. A cohort of healthy women, matched in age, underwent two chest CTs. EAT was quantified on chest CT using semi-automated software. CTX was defined as a > 10% reduction in left ventricular ejection fraction (LVEF) from baseline, with an absolute value of < 53%.
    UNASSIGNED: A total of 41 BC patients were included in the study, with 23 patients in the AC-T group and 18 patients in the AC-TH group. Additionally, 22 healthy females were included as the normal group. None of the BC patients developed CTX after chemotherapy. The age did not differ significantly between the normal group and the AC-T group (p = 0.341) or the AC-TH group (p = 0.853). Similarly, the body mass index (BMI) of the normal group was comparable to that of the AC-T group (p = 0.377, 0.346) and the AC-TH group (p = 0.148, 0.119) before and after chemotherapy. The EAT volume index (mL/kg/ m 2 ) was significantly higher in both the AC-T group (5.11 ± 1.85 vs. 4.34 ± 1.55, p < 0.001) and the AC-TH group (4.53 ± 1.61 vs. 3.48 ± 1.62, p < 0.001) at T1 compared with T0. In addition, both the AC-T group (-72.95 ± 5.01 vs. -71.22 ± 3.91, p = 0.005) and the AC-TH group (-72.55 ± 5.27 vs. -68.20 ± 5.98, p < 0.001) exhibited a significant decrease in EAT radiodensity (HU) at T1 compared to T0. However, there was no significant difference observed in the normal group. At T0, no difference was seen in EAT volume index (4.34 ± 1.55 vs. 3.48 ± 1.62, p = 0.090) and radiodensity (-71.22 ± 3.91 vs. -68.20 ± 5.98, p = 0.059) between the AC-T and AC-TH groups. Similarly, at T1, there was still no significant difference observed in the EAT volume index (-5.11 ± 1.85 vs. 4.53 ± 1.61, p = 0.308) and radiodensity (-72.95 ± 5.00 vs. -72.54 ± 5.27, p = 0.802) between the two groups.
    UNASSIGNED: BC patients who underwent AC-T and AC-TH adjuvant chemotherapy protocols demonstrated a significant rise in the volume index of EAT, along with a substantial reduction in its radiodensity post-chemotherapy. These findings indicate that alterations in EAT could potentially aid in identifying cardiac complications caused by chemotherapeutic agents and remind clinicians to focus on changes in EAT after adjuvant chemotherapy in BC patients to prevent the practical occurrence of CTX.
    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
    大约20%的乳腺癌患者HER2阳性。当前治疗的功效受到对曲妥珠单抗的原发性和继发性耐药性的限制。tRNA衍生片段(tRF)在各种癌症中显示出至关重要的调节作用。这项研究旨在评估tRF-27在调节HER2阳性乳腺癌对曲妥珠单抗的耐药性中的作用。tRF-27在曲妥珠单抗耐药细胞中高表达,其表达水平可以预测曲妥珠单抗的耐药性。tRF-27的高表达促进曲妥珠单抗暴露的细胞的生长和增殖。进行RNA下拉测定和质谱以鉴定RasGTP酶激活蛋白结合蛋白1和2(G3BP)(tRF-27靶向的两种蛋白质);RNA免疫沉淀(RIP)以确认其结合;共免疫沉淀(共IP)和RNA下拉测定以确定G3BP和tRF-27之间的结合域。tRF-27通过特定序列与G3BP的核转运因子2样结构域(NTF2结构域)结合。tRF-27依赖于G3BP和NTF2结构域来增加曲妥珠单抗耐受性。tRF-27与溶酶体相关膜蛋白1(LAMP1)竞争NTF2结构域,从而抑制G3BP和结节性硬化症(TSC)的溶酶体定位。tRF-27的过表达抑制了TSCs的磷酸化,并促进了雷帕霉素复合物1(MTORC1)的机制靶标的激活,以增强细胞增殖并诱导HER2阳性乳腺癌对曲妥珠单抗的抗性。
    About 20% of breast cancer patients are positive for HER2. The efficacy of current treatments is limited by primary and secondary resistance to trastuzumab. tRNA-derived fragments (tRFs) have shown crucial regulatory roles in various cancers. This study aimed to evaluate the role of tRF-27 in regulating the resistance of HER2-positive breast cancer against trastuzumab. tRF-27 was highly expressed in trastuzumab-resistant cells, and its expression level could predict the resistance to trastuzumab. High expression of tRF-27 promoted the growth and proliferation of trastuzumab-exposed cells. RNA-pulldown assay and mass spectrometry were performed to identify Ras GTPase-activating protein-binding proteins 1 and 2 (G3BPs) (two proteins targeted by tRF-27); RNA-immunoprecipitation (RIP) to confirm their bindings; co-immunoprecipitation (co-IP) and RNA-pulldown assay to determine the binding domains between G3BPs and tRF-27.tRF-27 bound to the nuclear transport factor 2 like domain(NTF2 domain) of G3BPs through a specific sequence. tRF-27 relied on G3BPs and NTF2 domain to increase trastuzumab tolerance. tRF-27 competed with lysosomal associated membrane protein 1(LAMP1) for NTF2 domain, thereby inhibiting lysosomal localization of G3BPs and tuberous sclerosis complex (TSC). Overexpression of tRF-27 inhibited phosphorylation of TSCs and promoted the activation of mechanistic target of rapamycin complex 1(MTORC1) to enhance cell proliferation and entice the resistance of HER2-positive breast cancer against trastuzumab.
    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
    乳腺癌(BC)是女性中普遍存在的恶性肿瘤,在所有BC的20-30%中观察到HER2过表达,从而作为受影响个体不利结果的预后指标。有必要建立创新的治疗方案,以扩大可用于管理HER2阳性BC的治疗替代方案。在这项研究中,我们报告了一例HER2阳性BC,在我们部门使用三种靶向药物的组合进行管理(曲妥珠单抗,帕妥珠单抗和吡罗替尼)以及化疗。治疗导致病理完全缓解(pCR),并观察到耐受性良好,无明显不良反应。因此,吡罗替尼和双重HER2阻断联合治疗有望作为局部晚期HER2阳性BC的新辅助治疗,在手术中实现pCR.然而,这一结论需要通过包含更多患者人群的精心设计的临床研究进行进一步验证.
    Breast cancer (BC) is the prevailing malignancy among women, with HER2 overexpression observed in 20-30 % of all BC, thereby serving as a prognostic indicator for unfavorable outcomes in affected individuals. There is a necessity to establish innovative treatment protocols to expand the therapeutic alternatives accessible for managing HER2-positive BC. In this study, we report a case of HER2-positive BC that was managed in our department using a combination of three targeted drugs (Trastuzumab, Pertuzumab and Pyrotinib) along with chemotherapy. The treatment resulted in a pathological complete response (pCR) and was observed to be well-tolerated, without any significant adverse reactions. Hence, the combination of Pyrotinib and Dual HER2 blockade treatment shows promise as a neoadjuvant therapy for locally advanced HER2-positive BC to achieve a pCR in surgery. Nevertheless, this conclusion necessitates additional validation via meticulously designed clinical research investigations encompassing larger patient populations.
    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
    目的:对靶向治疗的耐药是癌症治疗的关键障碍之一。在HER2+癌症的治疗中经常发展对曲妥珠单抗的抗性。蛋白酪氨酸磷酸酶(PTP)在曲妥珠单抗耐药性中的作用尚不清楚。在这项研究中,我们的目标是确定影响曲妥珠单抗耐药的关键PTP,并设计一种新的对抗策略.
    方法:使用四个公共数据集来筛选与HER2+乳腺癌患者曲妥珠单抗反应性相关的候选PTP。酪氨酸激酶(TK)阵列用于鉴定与蛋白酪氨酸磷酸受体O型(PTPRO)增强的曲妥珠单抗敏感性相关的激酶。在细胞模型中,测试了曲妥珠单抗缀合的二氧化硅纳米颗粒中的小激活RNA(saRNA)的PTPRO上调和抗性缓解的功效,转基因小鼠模型,和人类癌细胞系来源的异种移植模型。
    结果:PTPRO被确定为影响曲妥珠单抗反应性和患者生存的关键PTP。PTPRO去磷酸化了几个TK,包括以前被忽视的底物ERBB3,从而抑制与耐药性相关的多个致癌途径。值得注意的是,PTPRO,以前被认为是不可吸毒的,“被负载saRNA的纳米颗粒有效地上调。上调的PTPRO同时抑制ERBB3、ERBB2和下游SRC信号通路,从而抵消曲妥珠单抗耐药。
    结论:抗体缀合的saRNA代表了靶向“不可药物”PTP的创新方法。
    OBJECTIVE: Resistance to targeted therapy is one of the critical obstacles in cancer management. Resistance to trastuzumab frequently develops in the treatment for HER2+ cancers. The role of protein tyrosine phosphatases (PTPs) in trastuzumab resistance is not well understood. In this study, we aim to identify pivotal PTPs affecting trastuzumab resistance and devise a novel counteracting strategy.
    METHODS: Four public datasets were used to screen PTP candidates in relation to trastuzumab responsiveness in HER2+ breast cancer. Tyrosine kinase (TK) arrays were used to identify kinases that linked to protein tyrosine phosphate receptor type O (PTPRO)-enhanced trastuzumab sensitivity. The efficacy of small activating RNA (saRNA) in trastuzumab-conjugated silica nanoparticles was tested for PTPRO upregulation and resistance mitigation in cell models, a transgenic mouse model, and human cancer cell line-derived xenograft models.
    RESULTS: PTPRO was identified as the key PTP which influences trastuzumab responsiveness and patient survival. PTPRO de-phosphorated several TKs, including the previously overlooked substrate ERBB3, thereby inhibiting multiple oncogenic pathways associated with drug resistance. Notably, PTPRO, previously deemed \"undruggable,\" was effectively upregulated by saRNA-loaded nanoparticles. The upregulated PTPRO simultaneously inhibited ERBB3, ERBB2, and downstream SRC signaling pathways, thereby counteracting trastuzumab resistance.
    CONCLUSIONS: Antibody-conjugated saRNA represents an innovative approach for targeting \"undruggable\" PTPs.
    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)已证明在治疗各种癌症方面有效,特别是在靶向人类表皮生长因子受体2(HER2)阳性乳腺癌方面表现出特异性。3期临床试验的最新进展扩大了目前对ADC的理解,尤其是曲妥珠单抗deruxtecan,用于治疗其他表达HER2的恶性肿瘤。这种知识的扩展导致美国食品和药物管理局批准曲妥珠单抗deruxtecan治疗HER2阳性和低HER2乳腺癌,HER2阳性胃癌,和HER2突变型非小细胞肺癌。同时在肿瘤学中使用越来越多的ADC,卫生保健专业人员越来越关注间质性肺病或肺炎(ILD/p)的发病率上升,与抗HER2ADC治疗相关。关于抗HER2ADC的研究报告了不同的ILD/p死亡率。因此,在接受抗HER2ADC治疗的患者中,制定ILD/p的诊断和治疗指南至关重要.为此,我们召集了一个由中国专家组成的小组,以制定一项战略方法,用于识别和管理抗HER2ADC治疗患者的ILD/p.本报告提出专家小组的意见和建议,旨在指导临床实践中抗HER2ADC治疗诱导的ILD/p的管理。
    Antibody-drug conjugates (ADCs) have demonstrated effectiveness in treating various cancers, particularly exhibiting specificity in targeting human epidermal growth factor receptor 2 (HER2)-positive breast cancer. Recent advancements in phase 3 clinical trials have broadened current understanding of ADCs, especially trastuzumab deruxtecan, in treating other HER2-expressing malignancies. This expansion of knowledge has led to the US Food and Drug Administration\'s approval of trastuzumab deruxtecan for HER2-positive and HER2-low breast cancer, HER2-positive gastric cancer, and HER2-mutant nonsmall cell lung cancer. Concurrent with the increasing use of ADCs in oncology, there is growing concern among health care professionals regarding the rise in the incidence of interstitial lung disease or pneumonitis (ILD/p), which is associated with anti-HER2 ADC therapy. Studies on anti-HER2 ADCs have reported varying ILD/p mortality rates. Consequently, it is crucial to establish guidelines for the diagnosis and management of ILD/p in patients receiving anti-HER2 ADC therapy. To this end, a panel of Chinese experts was convened to formulate a strategic approach for the identification and management of ILD/p in patients treated with anti-HER2 ADC therapy. This report presents the expert panel\'s opinions and recommendations, which are intended to guide the management of ILD/p induced by anti-HER2 ADC therapy in clinical practice.
    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:Inetetamab是国内首个研发的创新抗HER2单克隆抗体,在HER2阳性晚期乳腺癌中被证明是有效和安全的。然而,其在HER2阳性局部晚期乳腺癌(LABC)新辅助治疗中的有效性和安全性仍有待验证.
    方法:这项前瞻性队列研究旨在评估伊奈他单抗联合帕妥珠单抗的疗效和安全性。紫杉烷,和卡铂(TCbIP)在HER2阳性LABC的新辅助治疗中,将其与接受TCbHP方案治疗的患者的数据进行比较(曲妥珠单抗联合帕妥珠单抗,紫杉烷,和卡铂)使用倾向评分匹配(PSM)。主要终点是总病理完全缓解(tpCR)。不良事件(AE),客观反应率(ORR),接近pCR是关键的次要终点。
    结果:44例临床阶段IIA-IIICHER2阳性LABC患者被前瞻性纳入并接受TCbIP方案治疗。28例完成手术的患者的tpCR率为60.7%,与PSM中的TCbHP组相当并稍高(60.7%与53.6%,P=0.510)。ORR为96.4%,DCR达到100.0%。最常见的≥3级AE是中性粒细胞减少症(21.4%与11.9%,P=0.350)。未观察到左心室射血分数显著降低,无患者因不良事件退出治疗。
    结论:TCbIP新辅助治疗在HER2阳性LABC患者中显示出良好的疗效和安全性,可能是新辅助治疗的另一种有希望的选择。
    背景:NCT05749016(注册日期:2021年11月1日)。
    BACKGROUND: Inetetamab is the first domestically developed innovative anti-HER2 monoclonal antibody in China, proven effective and safe in HER2-positive advanced breast cancer. However, its efficacy and safety in neoadjuvant treatment of HER2-positive locally advanced breast cancer (LABC) remain to be validated.
    METHODS: This prospective cohort study aimed to evaluate the efficacy and safety of inetetamab combined with pertuzumab, taxanes, and carboplatin (TCbIP) in neoadjuvant therapy for HER2-positive LABC, comparing it to data from patients treated with the TCbHP regimen (trastuzumab combined with pertuzumab, taxanes, and carboplatin) using propensity score matching (PSM). The primary endpoint was total pathological complete response (tpCR). Adverse events (AEs), objective response rate (ORR), and near-pCR were key secondary endpoints.
    RESULTS: Forty-four patients with clinical stage IIA-IIIC HER2-positive LABC were prospectively enrolled and treated with the TCbIP regimen. The tpCR rate among 28 patients who completed surgery was 60.7%, comparable to and slightly higher than the TCbHP group in PSM (60.7% vs. 53.6%, P = 0.510). The ORR was 96.4%, and the DCR reached 100.0%. The most common ≥ grade 3 AE was neutropenia (21.4% vs. 11.9%, P = 0.350). No significant reduction in left ventricular ejection fraction was observed, and no patient withdrew from treatment due to AEs.
    CONCLUSIONS: Neoadjuvant therapy with TCbIP showed good efficacy and safety in patients with HER2-positive LABC and might be another promising option for neoadjuvant treatment.
    BACKGROUND: NCT05749016 (registration date: Nov 01, 2021).
    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
    对HER2靶向治疗的抗性是HER2+乳腺癌(BC)患者治疗失败的主要原因。鉴于免疫微环境在肿瘤发展中的关键作用,缺乏一个理想的预后模型来完全解释免疫浸润.在这项研究中,进行WGCNA分析以发现免疫相关信号与HER2BC预后之间的关系。赫赛汀抗性BC细胞系建立后,对GSE76360队列的耐药细胞系转录谱和RNA测序数据进行了候选基因分析.通过Cox回归分析来自TCGA数据库的85个HER2+BC样本,XGBoost和Lasso算法推广可靠的免疫相关预后指数(IRPI)。通过多种算法进一步分析了IRPI特征与肿瘤微环境之间的相关性。包括单细胞RNA测序数据分析。高IRPI患者肿瘤免疫微环境抑制,预后较差。证实了在赫赛汀抗性HER2+BC中由IRPI指示的I型干扰素信号传导的抑制。并且我们阐明了cGAS-STING途径的抑制是具有高IRPI的赫赛汀抗性BC中免疫逃逸的关键决定因素。STING激动剂和DS-8201的组合可以作为赫赛汀抗性HER2+BC的新策略。
    Resistance to HER2-targeted therapy is the major cause of treatment failure in patients with HER2+ breast cancer (BC). Given the key role of immune microenvironment in tumor development, there is a lack of an ideal prognostic model that fully accounts for immune infiltration. In this study, WGCNA analysis was performed to discover the relationship between immune-related signaling and prognosis of HER2+ BC. After Herceptin-resistant BC cell lines established, transcriptional profiles of resistant cell line and RNA-sequencing data from GSE76360 cohort were analyzed for candidate genes. 85 samples of HER2+ BC from TCGA database were analyzed by the Cox regression, XGBoost and Lasso algorithm to generalize a credible immune-related prognostic index (IRPI). Correlations between the IRPI signature and tumor microenvironment were further analyzed by multiple algorithms, including single-cell RNA sequencing data analysis. Patients with high IRPI had suppressive tumor immune microenvironment and worse prognosis. The suppression of type I interferon signaling indicated by the IRPI in Herceptin-resistant HER2+ BC was validated. And we elucidated that the suppression of cGAS-STING pathway is the key determinant underlying immune escape in Herceptin-resistant BC with high IRPI. A combination of STING agonist and DS-8201 could serve as a new strategy for Herceptin-resistant HER2+ BC.
    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
    胃癌是世界范围内最常见的癌症之一,和人类表皮生长因子受体2(HER2)阳性病例约占总病例的20%。目前,曲妥珠单抗+化疗是HER2阳性晚期胃癌患者推荐的一线治疗方法,并且该组合在HER2靶向治疗中表现出明确的疗效。然而,治疗过程中耐药性的出现大大降低了其有效性;因此,研究潜在的耐药机制势在必行.在本评论文章中,我们全面介绍了HER2阳性胃癌病例对曲妥珠单抗耐药的多种机制,旨在为纠正与曲妥珠单抗耐药相关的问题和制定后续治疗策略提供见解。
    Gastric cancer is one of the most prevalent cancers worldwide, and human epidermal growth factor receptor 2 (HER2)-positive cases account for approximately 20% of the total cases. Currently, trastuzumab + chemotherapy is the recommended first-line treatment for patients with HER2-positive advanced gastric cancer, and the combination has exhibited definite efficacy in HER2-targeted therapy. However, the emergence of drug resistance during treatment considerably reduces its effectiveness; thus, it is imperative to investigate the potential mechanisms underlying resistance. In the present review article, we comprehensively introduce multiple mechanisms underlying resistance to trastuzumab in HER2-positive gastric cancer cases, aiming to provide insights for rectifying issues associated with resistance to trastuzumab and devising subsequent treatment strategies.
    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:曲妥珠单抗和帕妥珠单抗组合已被批准用于治疗HER2阳性转移性乳腺癌患者。然而,曲妥珠单抗和帕妥珠单抗联合治疗HER2阳性转移性胃癌患者的总生存期没有改善.
    方法:我们开发了一种新的HER2靶向单克隆抗体,HLX22,靶向HER2亚结构域IV作为曲妥珠单抗,但具有非重叠表位。我们检查了这种新型HER2抗体在胃细胞系和细胞系来源的异种移植物(CDX)和患者来源的异种移植物(PDX)模型中的抗肿瘤作用。
    结果:HLX22与HLX02(曲妥珠单抗生物仿制药)联合诱导HER2/HER2同源二聚体和HER2/EGFR异源二聚体内化的增强,最终导致涉及STAT3,P70S6和AKT的信号转导减少;FGF-FGFR-PI3K-MTOR的基因表达,EGF-EGFR-RAS,TGF-β-SMAD,PLCG和细胞周期进展相关的途径,有利于肿瘤的发展,扩散,programming,胃癌细胞系NCI-N87的迁移和存活也降低。这些不同但互补的作用有助于HLX22和HLX02组合在胃癌细胞系中的协同抗肿瘤功效,CDX和PDX。此外,HLX22与HLX02的组合在体外和体内表现出比HLX02和HLX11(潜在的帕妥珠单抗生物类似物)组合治疗更强的抗肿瘤功效。
    结论:这些结果表明,靶向HER2亚结构域IV的非竞争性抗体HLX22和HLX02一起应用可能对目前对曲妥珠单抗治疗反应欠佳的胃癌患者具有实质性益处。
    BACKGROUND: Trastuzumab and pertuzumab combination has been approved for the treatment of patients with HER2-positive metastatic breast cancer. However, trastuzumab and pertuzumab combination did not show improvement in overall survival in patients with HER2-positive metastatic gastric cancer.
    METHODS: We developed a new HER2-targeted monoclonal antibody, HLX22, targeting HER2 subdomain IV as trastuzumab but with non-overlapping epitopes. We examined the antitumor effects of this novel HER2-antibody in gastric cell lines and cell line-derived xenograft (CDX) and patient-derived xenograft (PDX) models.
    RESULTS: HLX22 in combination with HLX02 (trastuzumab biosimilar) induced enhancement of HER2/HER2 homodimers and HER2/EGFR heterodimers internalization, which ultimately led to the reduction in signal transductions involving STAT3, P70 S6, and AKT; gene expressions of FGF-FGFR-PI3K-MTOR, EGF-EGFR-RAS, TGF-β-SMAD, PLCG and cell cycle progression related pathways that favor tumor development, proliferation, progression, migration and survival in gastric cancer cell line NCI-N87 were also reduced. These differing but complementary actions contributed to the synergistic antitumor efficacy of the HLX22 and HLX02 combination in gastric cancer cell lines, CDX and PDX. In addition, HLX22 in combination with HLX02 demonstrated stronger antitumor efficacy than HLX02 and HLX11 (a potential pertuzumab biosimilar) combination treatment both in vitro and in vivo.
    CONCLUSIONS: These results suggested that the application of non-competing antibodies HLX22 and HLX02 targeting HER2 subdomain IV together may be of substantial benefit to gastric cancer patients who currently respond suboptimal to trastuzumab therapy.
    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
    嵌合抗原受体(CAR)T细胞疗法在治疗恶性肿瘤方面显示出希望。然而,使用人表皮生长因子受体2(HER2)CAR-T细胞具有严重毒性的风险,包括细胞因子释放综合征,由于他们对HER2的“上靶肿瘤外”识别。增强HER2CAR的质量和功能可以大大提高CAR-T细胞的治疗潜力。在这项研究中,我们开发了一种新型的抗HER2单克隆抗体,Ab8,其靶向HER2的结构域III,不同于曲妥珠单抗的结构域IV识别。尽管两种抗HER2mAb诱导了相似水平的抗体依赖性细胞毒性,基于曲妥珠单抗的CAR-T细胞对HER2阳性癌细胞表现出有效的抗肿瘤活性。总之,我们的研究结果提供了科学证据,即抗体对近侧膜结构域的识别可促进HER2特异性CAR-T细胞的抗肿瘤反应.
    Chimeric antigen receptor (CAR) T cell therapy shows promise in treating malignant tumors. However, the use of human epidermal growth factor receptor-2 (HER2) CAR-T cells carries the risk of severe toxicity, including cytokine release syndrome, due to their \"on-target off-tumor\" recognition of HER2. Enhancing the quality and functionality of HER2 CARs could greatly improve the therapeutic potential of CAR-T cells. In this study, we developed a novel anti-HER2 monoclonal antibody, Ab8, which targets domain III of HER2, distinct from the domain IV recognition of trastuzumab. Although two anti-HER2 mAbs induced similar levels of antibody-dependent cellular cytotoxicity, trastuzumab-based CAR-T cells exhibited potent antitumor activity against HER2-positive cancer cells. In conclusion, our findings provide scientific evidence that antibody recognition of the membrane-proximal domain promotes the anti-tumor response of HER2-specific CAR-T cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号