trastuzumab deruxtecan

曲妥珠单抗 deruxtecan
  • 文章类型: Journal Article
    背景:曲妥珠单抗deruxtecan(T-DXd)被批准用于人类表皮生长因子受体2(HER2)阳性和HER2低的晚期乳腺癌(ABC)。T-DXd在稳定或活跃的脑转移瘤(BMs)的HER2阳性ABC患者中显示出令人鼓舞的颅内活性;然而,其在HER2-低ABC伴BMs患者中的疗效尚不明确.
    方法:DEBBRAH是单臂,五组,II期研究评估来自HER2阳性和HER2低ABC的中枢神经系统受累患者的T-DXd。这里,我们报告了严重预处理HER2低ABC和活动性BM的患者的结果,纳入第2组(n=6,无症状未治疗的BMs)和第4组(n=6,局部治疗后进展的BMs).患者每21天一次静脉注射5.4mg/kgT-DXd。主要终点为两组神经肿瘤脑转移瘤(RANO-BM)的颅内客观反应率。
    结果:队列2和队列4中每个RANO-BM的颅内客观缓解率分别为50.0%[3/6例患者;95%置信区间(CI)11.8%至88.2%]和33.3%[2/6例患者;95%CI4.3%至77.7%;P=0.033(单侧)]。所有反应者都有部分反应。颅内反应的中位时间为2.3个月(范围,1.5-4.0个月),颅内反应的中位持续时间为7.2个月(范围,2.8-16.8个月)。根据RECISTv.1.1的无进展生存期中位数为5.4个月(95%CI4.1-10.0个月)。所有患者均发生因治疗引起的不良事件(16.7%为3级)。3例患者(25.0%)有1级间质性肺病/肺炎。
    结论:T-DXd在有活动性BMs的HER2低ABC预处理患者中表现出良好的颅内活性。需要进一步的研究在更大的队列中验证这些结果。该试验已在ClinicalTrials.gov注册,NCT04420598。
    BACKGROUND: Trastuzumab deruxtecan (T-DXd) is approved for human epidermal growth factor receptor 2 (HER2)-positive and HER2-low advanced breast cancer (ABC). T-DXd has shown encouraging intracranial activity in HER2-positive ABC patients with stable or active brain metastases (BMs); however, its efficacy in patients with HER2-low ABC with BMs is not well established yet.
    METHODS: DEBBRAH is a single-arm, five-cohort, phase II study evaluating T-DXd in patients with central nervous system involvement from HER2-positive and HER2-low ABC. Here, we report results from patients with heavily pretreated HER2-low ABC and active BMs, enrolled in cohorts 2 (n = 6, asymptomatic untreated BMs) and 4 (n = 6, progressing BMs after local therapy). Patients received 5.4 mg/kg T-DXd intravenously once every 21 days. The primary endpoint was intracranial objective response rate per Response Assessment in Neuro-Oncology Brain Metastases (RANO-BM) for both cohorts.
    RESULTS: Intracranial objective response rate per RANO-BM was 50.0% [3/6 patients; 95% confidence interval (CI) 11.8% to 88.2%] and 33.3% [2/6 patients; 95% CI 4.3% to 77.7%; P = 0.033 (one-sided)] in cohorts 2 and 4, respectively. All responders had partial responses. Median time to intracranial response was 2.3 months (range, 1.5-4.0 months) and median duration of intracranial response was 7.2 months (range, 2.8-16.8 months). Median progression-free survival per RECIST v.1.1. was 5.4 months (95% CI 4.1-10.0 months). Treatment-emergent adverse events occurred in all patients included (16.7% grade 3). Three patients (25.0%) had grade 1 interstitial lung disease/pneumonitis.
    CONCLUSIONS: T-DXd demonstrated promising intracranial activity in pretreated HER2-low ABC patients with active BMs. Further studies are needed to validate these results in larger cohorts. This trial is registered with ClinicalTrials.gov, NCT04420598.
    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:随着曲妥珠单抗deruxtecan(T-DXd)的广泛应用,HER2低的乳腺癌患者的生存率显著提高.然而,对T-DXd的耐药性仍然存在于一部分患者中,分子机制尚不清楚。
    方法:进行体内shRNA慢病毒文库功能筛选以鉴定介导T-DXd抗性的潜在环状RNA(crRNA)。RNA下拉,质谱,RNA免疫沉淀,并进行了免疫共沉淀测定以研究其分子机制。使用C11-BODIPY检测铁凋亡,Liperfluo,铁橙染色,谷胱甘肽定量,丙二醛定量,和透射电子显微镜。分子对接,虚拟筛选,和患者来源的异种移植(PDX)模型用于验证治疗药物.
    结果:VDAC3衍生的crRNA(crVDAC3)在功能性shRNA文库筛选中排名第一。敲除crVDAC3增加了低HER2乳腺癌细胞对T-DXd治疗的敏感性。进一步的机制研究表明,crVDAC3特异性结合HSPB1蛋白并抑制其泛素化降解,导致细胞内积累和HSPB1蛋白水平升高。值得注意的是,抑制crVDAC3可显着增加过量的ROS水平和不稳定的铁池积累。抑制crVDAC3通过降低HSPB1表达诱导乳腺癌细胞铁凋亡,从而介导T-DXd抗性。通过虚拟筛选和实验验证,我们确定paritaprevir可以有效地结合crVDAC3并阻止其与HSPB1蛋白的相互作用,从而增加HSPB1蛋白的泛素化降解以克服T-DXd抗性。最后,在HER2低PDX模型中,我们验证了paritaprevir增强T-DXd的治疗效果.
    结论:这一发现揭示了低HER2乳腺癌T-DXd耐药的分子机制。我们的研究提供了一种通过抑制crVDAC3与HSPB1蛋白之间的相互作用来克服T-DXd抗性的新策略。
    OBJECTIVE: With the wide application of trastuzumab deruxtecan (T-DXd), the survival of HER2-low breast cancer patients is dramatically improved. However, resistance to T-DXd still exists in a subset of patients, and the molecular mechanism remains unclear.
    METHODS: An in vivo shRNA lentiviral library functional screening was performed to identify potential circular RNA (crRNA) that mediates T-DXd resistance. RNA pull-down, mass spectrometry, RNA immunoprecipitation, and co-immunoprecipitation assays were conducted to investigate the molecular mechanism. Ferroptosis was detected using C11-BODIPY, Liperfluo, FerroOrange staining, glutathione quantification, malondialdehyde quantification, and transmission electron microscopy. Molecular docking, virtual screening, and patient-derived xenograft (PDX) models were used to validate therapeutic agents.
    RESULTS: VDAC3-derived crRNA (crVDAC3) ranked first in functional shRNA library screening. Knockdown of crVDAC3 increased the sensitivity of HER2-low breast cancer cells to T-DXd treatment. Further mechanistic research revealed that crVDAC3 specifically binds to HSPB1 protein and inhibits its ubiquitination degradation, leading to intracellular accumulation and increased levels of HSPB1 protein. Notably, suppression of crVDAC3 dramatically increases excessive ROS levels and labile iron pool accumulation. Inhibition of crVDAC3 induces ferroptosis in breast cancer cells by reducing HSPB1 expression, thereby mediating T-DXd resistance. Through virtual screening and experimental validation, we identified that paritaprevir could effectively bind to crVDAC3 and prevent its interaction with HSPB1 protein, thereby increasing ubiquitination degradation of HSPB1 protein to overcome T-DXd resistance. Finally, we validated the enhanced therapeutic efficacy of T-DXd by paritaprevir in a HER2-low PDX model.
    CONCLUSIONS: This finding reveals the molecular mechanisms underlying T-DXd resistance in HER2-low breast cancer. Our study provides a new strategy to overcome T-DXd resistance by inhibiting the interaction between crVDAC3 and HSPB1 protein.
    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:这项探索性汇总分析调查了基线时HER2阳性转移性乳腺癌(mBC)伴脑转移(BMs)患者曲妥珠单抗(T-DXd)与比较治疗的疗效和安全性,根据以前的局部治疗进行分类。
    方法:T-DXd数据来自DESTINY-Breast01/-02/-03。比较器数据,从接受医生选择治疗和曲妥珠单抗emtansine的患者,分别从DESTINY-Breast02和-03汇集。根据美国食品和药物管理局标准评估基线BM状态。终点包括根据RECISTv1.1进行的盲法独立中心检查(BICR)的颅内客观反应率(ORR;大脑完全或部分反应),颅内反应时间,颅内反应持续时间(DoR),BICR的中枢神经系统无进展生存期(CNS-PFS),总生存期(OS),和安全。
    结果:148例接受T-DXd治疗的患者和83例接受对照治疗的患者在基线时具有BMs。在那些接受T-DXd治疗的人中,治疗/稳定和未治疗/活动性BMs患者的颅内ORR分别为45.2%和45.5%,分别。治疗/稳定和未治疗/活动性BMs的患者颅内反应的中位(范围)时间为2.8个月(1.1-13.9个月)和1.5个月(1.2-13.7个月),分别。对于那些经过处理/稳定的BM,中位(95%CI)颅内DoR为12.3个月(9.1-17.9个月),对于那些未治疗/活跃的BMs,则为17.5个月(13.6-31.6个月)。中位(95%CI)CNS-PFS和OS为12.3个月(11.1-13.8个月),未达到(22.1个月-不可估计[NE]),和18.5个月(13.6-23.3个月)和30.2个月(21.3个月-NE)在那些未经处理/活性BMs,分别。43.2%的BMs患者和46.4%的T-DXd无BMs患者出现药物相关的TEAE等级≥3。
    结论:T-DXd在OS中显示有意义的颅内疗效和临床获益,在具有治疗/稳定和未治疗/活性BMs的HER2阳性mBC患者中具有可接受和可控制的安全性。
    BACKGROUND: This exploratory pooled analysis investigated the efficacy and safety of trastuzumab deruxtecan (T-DXd) versus comparator treatment in patients with HER2-positive metastatic breast cancer (mBC) with brain metastases (BMs) at baseline, categorized according to previous local treatment.
    METHODS: T-DXd data were pooled from DESTINY-Breast01/-02/-03. Comparator data, from patients receiving physician\'s choice therapy and trastuzumab emtansine, were pooled from DESTINY-Breast02 and -03, respectively. Baseline BM status was assessed according to US Food and Drug Administration criteria. Endpoints included intracranial objective response rate (ORR; complete or partial response in brain) per blinded independent central review (BICR) by RECIST v1.1, time to intracranial response, intracranial duration of response (DoR), central nervous system progression-free survival (CNS-PFS) by BICR, overall survival (OS), and safety.
    RESULTS: 148 patients who received T-DXd and 83 patients who received comparator treatment had BMs at baseline. In those who were treated with T-DXd, the intracranial ORR of patients with treated/stable and untreated/active BMs was 45.2% and 45.5%, respectively. The median (range) time to intracranial response was 2.8 months (1.1-13.9 months) and 1.5 months (1.2-13.7 months) in patients with treated/stable and untreated/active BMs, respectively. For those with treated/stable BMs, the median (95% CI) intracranial DoR was 12.3 months (9.1-17.9 months), and for those with untreated/active BMs it was 17.5 months (13.6-31.6 months). The median (95% CI) CNS-PFS and OS was 12.3 months (11.1-13.8 months) and not reached (22.1 months-not estimable [NE]) in those with treated/stable BMs, and 18.5 months (13.6-23.3 months) and 30.2 months (21.3 months-NE) in those with untreated/active BMs, respectively. Drug-related TEAEs grade ≥3 were experienced by 43.2% of patients with BMs and 46.4% without BMs with T-DXd.
    CONCLUSIONS: T-DXd demonstrated meaningful intracranial efficacy and clinical benefit in OS, with an acceptable and manageable safety profile in patients with HER2-positive mBC with treated/stable and untreated/active BMs.
    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
    UNASSIGNED: The third-generation antibody-drug conjugates (ADC), trastuzumab deruxtecan (T-DXd) and sacituzumab govitecan (SG), recently obtained approval for metastatic breast cancer treatment across various subtypes and therapeutic contexts.
    UNASSIGNED: This retrospective, multicentric study evaluated real-world tolerability, feasibility and efficacy in a pre-treated, real-world cohort at three major German breast cancer centers.
    UNASSIGNED: 125 patients treated with T-DXd or SG from November 2020 to June 2023 were included (T-DXd: 77 patients; SG: 48 patients). The median treatment duration was 6.0 months for T-DXd and 3.5 months for SG therapy, with a median follow-up duration of 10.4 months for T-DXd (95% CI: 8.4-11.6) and 11.8 months for SG (95% CI: 8.0-14.4). Severe neutropenia (CTC ≥ III°) occurred in 33.3% during SG therapy, with a numerical reduction observed following primary, prophylactic use of G-CSF. T-DXd-associated pneumonitis occurred in 8 out of 77 patients (10.4 %). Median progression-free survival (mPFS) was 8.6 months (95% CI: 5.8-12.4) with T-DXd (HER2+: 10.8; HER2-low: 4.7) and 4.9 months (95% CI: 2.8-6.3) with SG (TNBC 4.9; HR+/HER2-: not reached). Median overall survival (OS) was 23.8 months (95% CI: 16.1-not estimable) with T-DXd (HER2+: 27.1; HER2-low: not reached), and 12.4 months (95% CI: 8.7-not estimable) with SG therapy (TNBC: 12.4, HR+/HER2-: not reached). 95.7% of the protocol-specified, therapeutic dose was administered for T-DXd and 89.6% for SG.
    UNASSIGNED: Overall, this indicates good feasibility, tolerability, and effectiveness of ADC therapies in the real-world setting.
    UNASSIGNED: Trastuzumab-Deruxtecan (T-DXd) und Sacituzumab-Govitecan (SG) sind Antikörper-Wirkstoff-Konjugate (ADCs) der 3. Generation, die vor Kurzem zur Behandlung von metastatischem Brustkrebs über mehrere Subtypen hinweg und in verschiedenen therapeutischen Zusammenhängen zugelassen wurden.
    UNASSIGNED: Ziel dieser retrospektiven multizentrischen Studie war es, die Real-World-Daten über die Verträglichkeit, Umsetzbarkeit und Wirksamkeit dieser Wirkstoffe in einer vorbehandelten Real-World-Kohorte in 3 großen deutschen Brustkrebszentren zu bewerten.
    UNASSIGNED: Eingeschlossen wurden 125 Patientinnen, die zwischen November 2020 und Juni 2023 mit T-DXd oder SG behandelt wurden (T-DXd: 77 Patientinnen; SG: 48 Patientinnen). Die mediane Behandlungsdauer betrug 6,0 Monate für eine T-DXd- und 3,5 Monate für eine SG-Therapie mit einer medianen Nachbeobachtungszeit von 10,4 Monaten für T-DXd (95%-KI: 8,4–11,6) und 11,8 Monaten für SG (95%-KI: 8,0–14,4). 33,3% der Patientinnen entwickelten eine schwere Neutropenie (CTC ≥ III°) im Verlauf der SG-Therapie, wobei eine numerische Reduktion nach dem primären prophylaktischen Einsatz von G-CSF beobachtet wurde. Bei 8 von 77 Patientinnen (10,4 %) trat eine T-DXd-bedingte Pneumonitis auf. Das mediane progressionsfreie Überleben (mPFÜ) betrug 8,6 Monate (95%-KI: 5,8–12,4) mit T-DXd (HER2+: 10,8; HER2-low: 4,7) und 4,9 Monate (95%-KI: 2,8–6,3) mit SG (TNBC 4,9; HR+/HER2−: nicht erreicht). Das mediane Gesamtüberleben (GÜ) betrug 23,8 Monate (95%-KI: 16,1–nicht schätzbar) mit einer T-DXd-Therapie (HER2+: 27,1; HER2-low: nicht erreicht) und 12,4 Monate (95%-KI: 8,7–nicht schätzbar) mit einer SG-Therapie (TNBC: 12,4, HR+/HER2−: nicht erreicht). Verabreicht wurden 95,7% der im Protokoll vorgegebenen therapeutischen T-DXd-Dosis bzw. 89,6% der vorgegebenen SG-Dosis.
    UNASSIGNED: Insgesamt weisen die Daten auf eine gute Umsetzbarkeit, Wirksamkeit und Verträglichkeit von ADC-Therapien in einer realen Umgebung hin.
    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
    靶向表达人表皮生长因子受体2(HER2)的胃癌的治疗进展随着表达HER2的乳腺癌治疗的巨大成功,这促进了抗HER2药物适应症的扩展,不仅包括常规HER2阳性乳腺癌,而且HER2低和HER2超低亚组。低HER2胃癌的靶向性,然而,尚未建立。因此,需要进一步的研究来全面了解临床病理特征,特定的基因改变,和HER2低胃癌独特的肿瘤免疫微环境,并与HER2阳性或阴性胃癌进行比较。HER2的抗体-药物缀合物在使低HER2胃癌可靶向中起重要作用。在这种情况下,对新型抗HER2药物有更深入的了解,包括抗体-药物缀合物,双特异性T细胞接合抗体,这些药剂的组合,以及新形式的免疫调节剂也是必需的。不仅通过免疫组织化学/荧光原位杂交,而且通过评估使用DNA或RNA测序测量的ERRB2拷贝数增加或蛋白质过表达水平来重新定义和重新分类HER2状态可能有助于鉴定具有HER2表达肿瘤的群体,这些肿瘤理想地受益于抗HER2治疗。本论文回顾了最近的临床试验,特别关注低HER2胃癌以及基本/转化发现,并讨论了该独特亚组治疗的进一步治疗发展的观点。
    Therapeutic developments in the targeting of human epidermal growth factor receptor 2 (HER2)-expressing gastric cancer have followed the dramatic success of HER2-expressing breast cancer treatment, which has facilitated the expansion of indications for anti-HER2 agents to include not only conventional HER2-positive breast cancer, but also HER2-low and HER2-ultralow subgroups. The targetability of HER2-low gastric cancer, however, has yet to be established. Hence, further studies are needed to comprehensively understand the clinicopathological features, specific gene alterations, and distinct tumor immune microenvironment of HER2-low gastric cancer and compare them with those for HER2-positive or -negative gastric cancer. Antibody-drug conjugates for HER2 play an important role in making HER2-low gastric cancer targetable. In this context, a deeper understanding of the novel anti-HER2 agents, including antibody-drug conjugates, bispecific T-cell engager antibodies, and a combination of these agents, as well as new forms of immunomodulatory agents are also required. Redefining and re-categorizing HER2 status through not only immunohistochemistry/fluorescence in situ hybridization but also evaluating ERRB2 copy number gain or protein overexpression levels measured using DNA or RNA sequencing might be helpful for identifying populations with HER2-expressing tumors who would ideally benefit from anti-HER2 treatment. The current paper reviewed recent clinical trials, focusing particularly on HER2-low gastric cancer together with basic/translational findings, and discuss perspectives on further therapeutic development in the treatment of this distinct subgroup.
    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
    我们提供了一项关于曲妥珠单抗deruxtecan(T-DXd)在人表皮生长因子受体2阳性(HER2+)乳腺癌伴脑转移(BM)和/或软脑膜疾病(ROSET-BM)患者中的有效性的回顾性研究的最新结果(中位随访时间:20.4个月)。中位无进展生存期(PFS)为14.6个月。中位总生存期(OS)未达到(NR);24个月OS率为56.0%。亚组分析显示,分析活性BM患者的中位PFS为13.2个月,17.5个月的患者与软脑膜癌(LMC),分析稳定BM患者的NR(分析活跃BM患者的24个月PFS率,LMC,分析稳定的BM为32.7%,25.1%,和60.8%,分别)。LMC或分析稳定BM患者的分析活性BM和NR患者的中位OS为27.0个月(分析活性BM患者的24个月OS率,LMC,分析稳定的BM为52.0%,61.6%,71.6%,分别)。导致T-DXd停药的最常见不良事件是间质性肺病(ILD;23.1%);因ILD而停止T-DXd治疗的患者的ILD中位发病时间为5.3个月。T-DXd在严重预处理的HER2+转移性乳腺癌BM和LMC患者中具有有希望的有效性。ILD的发病率和中位发病时间与先前研究中日本亚组相似。
    We provide updated results (median follow-up duration: 20.4 months) of a retrospective study on the effectiveness of trastuzumab deruxtecan (T-DXd) in patients with human epidermal growth factor receptor 2-positive (HER2+) breast cancer with brain metastases (BM) and/or leptomeningeal disease (ROSET-BM). Median progression-free survival (PFS) was 14.6 months. Median overall survival (OS) was not reached (NR); 24-month OS rate was 56.0%. Subgroup analysis showed that median PFS was 13.2 months in patients with analytical active BM, 17.5 months in patients with leptomeningeal carcinomatosis (LMC), and NR in patients with analytical stable BM (24-month PFS rates in patients with analytical active BM, LMC, and analytical stable BM were 32.7%, 25.1%, and 60.8%, respectively). Median OS was 27.0 months in patients with analytical active BM and NR in patients with LMC or analytical stable BM (24-month OS rates in patients with analytical active BM, LMC, and analytical stable BM were 52.0%, 61.6%, and 71.6%, respectively). The most common adverse event leading to discontinuation of T-DXd was interstitial lung disease (ILD; 23.1%); median ILD onset time among patients who discontinued T-DXd treatment due to ILD was 5.3 months. T-DXd has promising effectiveness in heavily pre-treated HER2+ metastatic breast cancer patients with BM and LMC. The incidence and median onset time of ILD were similar to those of Japanese subgroups in previous studies.
    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: Case Reports
    在实体恶性肿瘤中,具有脑转移的乳腺癌占第二大数量的脑转移。尽管HER2靶向治疗取得了进展,50%的人表皮生长因子受体2阳性(HER2+)乳腺癌患者发生脑转移,并与不良预后相关。在这篇文章中,我们报道一例HER2+转移性乳腺癌患者发生脑转移,尽管使用曲妥珠单抗和帕妥珠单抗治疗后对颅外转移有持久的影响.患者在接受二次脑放疗和抗HER2药物多线治疗后接受曲妥珠单抗deruxtecan单药治疗时表现出颅内进展。如吡唑替尼,拉帕替尼,图卡替尼,和ado-曲妥珠单抗emtansine.然而,安洛替尼(抗血管生成药物)和曲妥珠单抗deruxtecan的给药导致颅内和颅外部分反应,并与可控的副作用相关.目前的情况表明,安洛替尼和曲妥珠单抗德鲁克替康的组合可能是HER2+乳腺癌脑转移患者的有希望的治疗选择。然而,需要进一步的研究来验证本研究结果.
    Breast cancer with brain metastasis accounts for the second largest number of brain metastases among solid malignancies. Despite advances in HER2-targeted therapy, 50% of patients with human epidermal growth factor receptor 2-positive (HER2+) breast cancer develop brain metastases and are associated with poor outcomes. In this article, we report the case of a patient with HER2+ metastatic breast cancer who developed brain metastases, despite experiencing a durable effect on extracranial metastases after treatment with trastuzumab and pertuzumab. The patient exhibited intracranial progression while receiving treatment with trastuzumab deruxtecan monotherapy after secondary brain radiotherapy and multiple lines of therapy with anti-HER2 agents, such as pyrotinib, lapatinib, tucatinib, and ado-trastuzumab emtansine. However, the administration of anlotinib (an antiangiogenesis medication) and trastuzumab deruxtecan resulted in intracranial and extracranial partial response and was linked to manageable side effects. The present case indicates that the combination of anlotinib and trastuzumab deruxtecan may be a promising treatment option for patients with HER2+ breast cancer with brain metastasis. Nevertheless, further studies are warranted to verify the present findings.
    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)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
    目的:新数据表明曲妥珠单抗deruxtecan(T-DXd)是HER2+乳腺癌脑转移的积极治疗方法。我们旨在表征T-DXd在一系列HER2改变的癌症的软脑膜转移(LM)治疗中的活性。
    方法:我们回顾了2020年7月至2023年12月的神经肿瘤学临床记录,以确定接受T-DXd治疗LM的患者。
    结果:在确定的18名患者中,6人患有HER2+乳腺癌,8人患有HER2低/阴性乳腺癌,2患有HER2+胃食管癌,2例患有HER2突变型非小细胞肺癌(NSCLC)。10/18(56%)患者通过CSF细胞学或循环肿瘤细胞(CTC)捕获在细胞学上证实了LM。使用EORTC/RANO-LM修订记分卡的MRI部分反应(PR)发生在4/6(67%)HER2乳腺LM患者中,2/8(25%)HER2低/阴性乳腺癌患者,和0/4(0%)的HER2胃食管癌或HER2突变型NSCLC患者。启动T-DXd后的中位总生存期为5.8个月。与HER2低/阴性乳腺癌和HER2改变的非乳腺癌患者相比,HER2+乳腺癌患者开始T-DXd后的生存期在数值上更长(13.9个月vs.5.2个月和4.6个月,分别)。Landmark分析显示,放射学LM对T-DXd有2.5个月反应的患者比无反应者的生存期长(14.2个月vs.2.6个月,HR0.18,95%CI0.05-0.63,p<0.05),T-DXd开始后3.5个月和4.5个月的界标分析显示出相似但不显著的趋势.
    结论:T-DXd在一部分患者中诱导LM反应,这种反应可能与生存期的延长有关。需要进行前瞻性试验以阐明T-DXd在治疗LM中的作用以及哪些患者最有可能受益。
    OBJECTIVE: Emerging data suggest that trastuzumab deruxtecan (T-DXd) is an active treatment for brain metastases from HER2 + breast cancer. We aimed to characterize the activity of T-DXd in the treatment of leptomeningeal metastases (LM) from a range of HER2-altered cancers.
    METHODS: We reviewed neuro-oncology clinic records between July 2020 and December 2023 to identify patients who received T-DXd to treat LM.
    RESULTS: Of 18 patients identified, 6 had HER2 + breast cancer, 8 had HER2-low/negative breast cancer, 2 had HER2 + gastroesophageal cancer, and 2 had HER2-mutant non-small cell lung cancer (NSCLC). 10/18 (56%) patients had cytologically confirmed LM by CSF cytology or circulating tumor cell (CTC) capture. A partial response (PR) on MRI using the EORTC/RANO-LM Revised-Scorecard occurred in 4/6 (67%) patients with HER2 + breast LM, 2/8 (25%) patients with HER2-low/negative breast cancer, and 0/4 (0%) patients with HER2 + gastroesophageal cancer or HER2-mutant NSCLC. Median overall survival after initiating T-DXd was 5.8 months. Survival after initiating T-DXd was numerically longer for HER2 + breast cancer patients compared with HER2-low/negative breast and HER2-altered non-breast cancer patients (13.9 months vs. 5.2 months and 4.6 months, respectively). Landmark analysis showed that patients with radiologic LM response to T-DXd by 2.5 months had longer survival than non-responders (14.2 months vs. 2.6 months, HR 0.18, 95% CI 0.05-0.63, p < 0.05), and landmark analyses at 3.5 and 4.5 months after starting T-DXd showed a similar but nonsignificant trend.
    CONCLUSIONS: T-DXd induces LM responses in a subset of patients, and such responses may be associated with prolongation of survival. Prospective trials are needed to clarify the role of T-DXd in treating LM and which patients are most likely to benefit.
    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
    人类表皮生长因子2(HER2)阳性乳腺癌(BC)占所有乳腺肿瘤的近20%。历史上,这些患者复发率高,预后差.HER2靶向单克隆抗体如曲妥珠单抗和帕妥珠单抗的出现改善了HER2阳性转移性BC的预后。最近,抗体-药物偶联物(ADC)现在正在重塑实体瘤的治疗模式,尤其是乳腺癌.曲舒珠单抗emtansine(T-DM1)是肿瘤学中开发的第一个ADC之一,并被批准用于HER2阳性转移性BC的治疗。在面对面的比较中,曲妥珠单抗deruxtecan(T-DXd)作为二线治疗击败T-DM1。ADC的功效被对这些试剂的获得性抗性的出现所抵消。在本文中,我们总结了T-DM1和T-DXd的作用机制和抗性,以及其临床疗效。此外,我们还讨论了解决ADC电阻的潜在策略。
    Human epidermal growth factor 2 (HER2)-positive breast cancer (BC) represents nearly 20% of all breast tumors. Historically, these patients had a high rate of relapse and dismal prognosis. The advent of HER2-targeting monoclonal antibodies such as trastuzumab followed by pertuzumab had improved the prognosis of HER2-positive metastatic BC. More recently, antibody-drug conjugates (ADCs) are now reshaping the treatment paradigm of solid tumors, especially breast cancer. Tratsuzumab emtansine (T-DM1) was one of the first ADC developed in oncology and was approved for the management of HER2-positive metastatic BC. In a head-to-head comparison, trastuzumab deruxtecan (T-DXd) defeated T-DM1 as a second-line treatment. The efficacy of ADCs is counterbalanced by the appearance of acquired resistance to these agents. In this paper, we summarize the mechanisms of action and resistance of T-DM1 and T-DXd, as well as their clinical efficacy. Additionally, we also discuss potential strategies for addressing resistance to ADC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号