Ado-Trastuzumab Emtansine

ado - 曲妥珠单抗 emtansine
  • 文章类型: English Abstract
    UNASSIGNED: HER-2 positive (+) breast cancer (BC) accounts for 20-25% of BC, it is more aggressive, and it has a lower survival rate. Since the approval of trastuzumab in 1998, other HER-2-targeted therapies such as pertuzumab and trastuzumab emtansine (TDM1) have been introduced, improving patient survival. However, cardiotoxicity is an adverse effect of these treatments.
    UNASSIGNED: To estimate the incidence of cardiotoxicity with trastuzumab, trastuzumab/pertuzumab, and TDM1 in women with HER-2 + BC treated over a 6-year period at the Hospital de Clínicas and the Hospital Departamental de Soriano.
    UNASSIGNED: Observational, descriptive, and retrospective study which included patients with HER-2 + BC treated with trastuzumab, trastuzumab/pertuzumab, or TDM1.
    UNASSIGNED: 81 patients were included, with a cardiotoxicity incidence of 23.4%. Cardiotoxicity was determined by a > 10% decrease in left ventricular ejection fraction (LVEF) (57.9%) and a LVEF < 50% evident during treatment (42.1%). Only 1 patient presented symptomatic heart failure. 63.1% of those who discontinued treatment due to cardiotoxicity managed to resume it. No relationship was evident between cardiovascular history or the administration regimen and the development of cardiotoxicity.
    UNASSIGNED: The study showed a cardiotoxicity incidence similar to the international one. Most did not present cardiac toxicity, and if they did, it was asymptomatic and reversible.
    UNASSIGNED: el cáncer de mama (CM) HER-2 positivo (+) representa el 20-25% de los CM, es más agresivo y tiene menor sobrevida. Desde la aprobación del trastuzumab en 1998, se han introducido otras terapias dirigidas al HER-2 como pertuzumab y trastuzumab emtansina (TDM1), con lo cual ha mejorado la supervivencia de las pacientes. Sin embargo, la cardiotoxicidad representa un efecto adverso de estos tratamientos.
    UNASSIGNED: estimar la incidencia de cardiotoxicidad con trastuzumab, trastuzumab/pertuzumab y TDM1 en mujeres con CM HER-2 +, tratadas en un periodo de 6 años en el Hospital de Clínicas y el Hospital Departamental de Soriano.
    UNASSIGNED: estudio observacional, descriptivo y retrospectivo que incluyó pacientes con CM HER-2 +, tratadas con trastuzumab, trastuzumab/pertuzumab o TDM1.
    UNASSIGNED: se incluyeron 81 pacientes. La incidencia de cardiotoxicidad fue del 23.4%. La cardiotoxicidad se determinó por una disminución > 10% de la fracción de ejección del venticulo izquierdo (FEVI) (57.9%) y por una FEVI < 50%, evidenciada durante el tratamiento (42.1%). Únicamente una paciente presentó insuficiencia cardiaca sintomática. El 63.1% de quienes suspendieron el tratamiento por cardiotoxicidad logró reanudarlo. No se evidenció una relación entre los antecedentes cardiovasculares ni con el esquema de administración y el desarrollo de cardiotoxicidad.
    UNASSIGNED: el estudio mostró una incidencia de cardiotoxicidad similar a la internacional. La mayoría no tuvo toxicidad cardiaca y si la hubo fue asintomática y reversible.
    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:HER2阳性乳腺癌脑转移(BCBM)的治疗选择仍然有限。我们先前报道了neratinib和neratinib-卡培他滨的中枢神经系统(CNS)活性。临床前数据表明,当联合给药时,neratinib可以克服对ado-曲妥珠单抗-emtansine(T-DM1)的耐药性。在TBCRC022的队列4中,我们检查了neratinib联合T-DM1对HER2阳性BCBM患者的疗效。
    方法:在这个多中心中,第二阶段研究,具有可测量的HER2阳性BCBM的患者在三个平行注册队列中接受了neratinib160mg每日加T-DM13.6mg/kg静脉内每21天一次(队列4A-先前未治疗的BCBM,队列4B和4C-BCBM在没有[4B]和[4C]之前暴露于T-DM1)的局部CNS定向治疗后进展。需要预防第1周期的腹泻。主要终点是按队列划分的神经肿瘤脑转移反应评估(RANO-BM)。还评估了总生存期(OS)和毒性。
    结果:在2018-2021年之间,有6、17和21名患者加入了队列4A,4B,和4C。由于累积缓慢,过早停止了注册。队列4A的CNS客观反应率,4B,4C为33.3%(95%置信区间[CI]:4.3-77.7%),35.3%(95%CI:14.2-61.7%),和28.6%(95%CI:11.3-52.2%),分别为38.1-50%,病情稳定≥6个月或有缓解。腹泻是最常见的3级毒性(22.7%)。中位OS为30.2个月(队列4A;95%CI:21.9,未达到[NR]),23.3个月(队列4B;95%CI:17.6,NR),和20.9个月(队列4C;95%CI:14.9,NR)。
    结论:我们观察到neratinib加T-DM1的颅内活动,包括那些先前有T-DM1暴露的患者,提示与neratinib的协同作用。我们的数据为HER2阳性BCBM患者基于neratinib的联合用药提供了更多证据。即使是那些被严重预处理的人。
    BACKGROUND: Treatment options for human epidermal growth factor receptor 2 (HER2)-positive breast cancer brain metastases (BCBMs) remain limited. We previously reported central nervous system (CNS) activity for neratinib and neratinib-capecitabine. Preclinical data suggest that neratinib may overcome resistance to ado-trastuzumab emtansine (T-DM1) when given in combination. In Translational Breast Cancer Research Consortium (TBCRC) 022\'s cohort 4, we examined the efficacy of neratinib plus T-DM1 in patients with HER2-positive BCBM.
    METHODS: In this multicenter, phase II study, patients with measurable HER2-positive BCBM received neratinib 160 mg daily plus T-DM1 3.6 mg/kg intravenously every 21 days in three parallel-enrolling cohorts [cohort 4A-previously untreated BCBM, cohorts 4B and 4C-BCBM progressing after local CNS-directed therapy without (4B) and with (4C) prior exposure to T-DM1]. Cycle 1 diarrheal prophylaxis was required. The primary endpoint was the Response Assessment in Neuro-Oncology-Brain Metastases (RANO-BM) by cohort. The overall survival (OS) and toxicity were also assessed.
    RESULTS: Between 2018 and 2021, 6, 17, and 21 patients enrolled in cohorts 4A, 4B, and 4C. Enrollment was stopped prematurely for slow accrual. The CNS objective response rate in cohorts 4A, 4B, and 4C was 33.3% [95% confidence interval (CI) 4.3% to 77.7%], 35.3% (95% CI 14.2% to 61.7%), and 28.6% (95% CI 11.3% to 52.2%), respectively; 38.1%-50% experienced stable disease for ≥6 months or response. Diarrhea was the most common grade 3 toxicity (22.7%). The median OS was 30.2 [cohort 4A; 95% CI 21.9-not reached (NR)], 23.3 (cohort 4B; 95% CI 17.6-NR), and 20.9 (cohort 4C; 95% CI 14.9-NR) months.
    CONCLUSIONS: We observed intracranial activity for neratinib plus T-DM1, including those with prior T-DM1 exposure, suggesting synergistic effects with neratinib. Our data provide additional evidence for neratinib-based combinations in patients with HER2-positive BCBM, even those who are heavily pretreated.
    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:在靶向治疗期间,HER2阳性乳腺癌总是失去HER2DNA扩增。相比之下,有趣的是,HER2蛋白可能丢失或获得。为了纵向和系统地了解HER2DNA/蛋白质化学计量的复杂/不一致变化,HER2DNA拷贝数和可溶性血液蛋白(aHER2/sHER2)平行测试,非侵入性(通过液体活检),在二维中,因此HER2-2D。
    方法:在使用抗体-药物偶联物(ADC)曲妥珠单抗-emtansine(T-DM1)对晚期HER2阳性乳腺癌患者(n=37)进行标准治疗之前和之后,通过数字PCR和ELISA评估aHER2和sHER2。
    结果:如预期,aHER2总是被T-DM1抑制,但这种损失令人惊讶地反映了sHER2增益,有时是相当大的实体,在大多数(30/37;81%)患者中。HER2致癌剂量的这种非正统分裂在两个代表性病例中得到了aHER2/sHER2动力学的支持。和免疫组织化学高状态,尽管4/5的T-DM1肿瘤后可再活检sHER2患者的拷贝数中性。此外,通过T-DM1体外处理的垂死的乳腺癌细胞系优先释放sHER2。最后,sHER2增加与比sHER2损失更长的PFS相关(平均PFS282天vs133天,95%CI[210-354]对[56-209],对数秩检验p=0.047),特别是当排除在T-DM1治疗期间出现循环HER2旁路改变的病例(n=11)时(平均PFS349vs139天,95%CI[255-444]vs[45-232],对数秩检验p=0.009)。
    结论:HER2增益是在肿瘤组织中适应性选择的,并通过sHER2增益在血液中概括。可能,在用裸抗体抗HER2治疗期间,增加致癌剂量对肿瘤有益,但在用强细胞毒性ADC如T-DM1治疗期间对宿主有利。在后一种情况下,HER2-gain肿瘤可以暂时保持在检查,直到替代致癌驱动因素,液体活检显示,绕过HER2。无论哪种解释,HER2-2D可能有助于定制/优先考虑抗HER2治疗,尤其是对低aHER2/低sHER2肿瘤有活性的ADC。
    背景:NCT05735392于2023年1月31日回顾性注册https://www。
    结果:gov/search?term=NCT05735392。
    BACKGROUND: During targeted treatment, HER2-positive breast cancers invariably lose HER2 DNA amplification. In contrast, and interestingly, HER2 proteins may be either lost or gained. To longitudinally and systematically appreciate complex/discordant changes in HER2 DNA/protein stoichiometry, HER2 DNA copy numbers and soluble blood proteins (aHER2/sHER2) were tested in parallel, non-invasively (by liquid biopsy), and in two-dimensions, hence HER2-2D.
    METHODS: aHER2 and sHER2 were assessed by digital PCR and ELISA before and after standard-of-care treatment of advanced HER2-positive breast cancer patients (n=37) with the antibody-drug conjugate (ADC) Trastuzumab-emtansine (T-DM1).
    RESULTS: As expected, aHER2 was invariably suppressed by T-DM1, but this loss was surprisingly mirrored by sHER2 gain, sometimes of considerable entity, in most (30/37; 81%) patients. This unorthodox split in HER2 oncogenic dosage was supported by reciprocal aHER2/sHER2 kinetics in two representative cases, and an immunohistochemistry-high status despite copy-number-neutrality in 4/5 available post-T-DM1 tumor re-biopsies from sHER2-gain patients. Moreover, sHER2 was preferentially released by dying breast cancer cell lines treated in vitro by T-DM1. Finally, sHER2 gain was associated with a longer PFS than sHER2 loss (mean PFS 282 vs 133 days, 95% CI [210-354] vs [56-209], log-rank test p=0.047), particularly when cases (n=11) developing circulating HER2-bypass alterations during T-DM1 treatment were excluded (mean PFS 349 vs 139 days, 95% CI [255-444] vs [45-232], log-rank test p=0.009).
    CONCLUSIONS: HER2 gain is adaptively selected in tumor tissues and recapitulated in blood by sHER2 gain. Possibly, an increased oncogenic dosage is beneficial to the tumor during anti-HER2 treatment with naked antibodies, but favorable to the host during treatment with a strongly cytotoxic ADC such as T-DM1. In the latter case, HER2-gain tumors may be kept transiently in check until alternative oncogenic drivers, revealed by liquid biopsy, bypass HER2. Whichever the interpretation, HER2-2D might help to tailor/prioritize anti-HER2 treatments, particularly ADCs active on aHER2-low/sHER2-low tumors.
    BACKGROUND: NCT05735392 retrospectively registered on January 31, 2023 https://www.
    RESULTS: gov/search?term=NCT05735392.
    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
    背景和目的:人表皮生长因子受体2(HER2)阳性,激素受体阳性(HR阳性)转移性乳腺癌(MBC)通常在后续系列中接受曲妥珠单抗emtansine(T-DM1)治疗.将内分泌治疗(ET)与T-DM1结合可以改善该亚型的治疗结果。因此,本研究旨在探讨T-DM1联合ET在HER2阳性和HR阳性MBC中的获益.这项研究是第一个研究ET与T-DM1结合的益处。材料和方法:本研究分析了2010年6月至2021年12月接受T-DM1治疗的HER2阳性和HR阳性MBC患者的医疗记录。根据患者是否同时接受ET和T-DM1分为两组。主要终点是确定无进展生存期(PFS),而次要终点是总生存期(OS),客观反应率,和治疗的安全性。结果:我们的分析检查了88例患者,其中32例(36.4%)接受T-DM1联合ET治疗。联合治疗显示中位数PFS显着改善(15.4vs.6.4个月;p=0.00004)和中位OS(35.0与23.1个月;p=0.026)与单独的T-DM1相比。组合组的ORR也较高(65.6%vs.29.3%;p=0.026)。与未接受帕妥珠单抗治疗的患者相比,先前接受帕妥珠单抗治疗的患者在T-DM1上的中位PFS降低(11.7vs.5.4个月,分别为;p<0.01)。与HER2+患者相比,T-DM1在HER23+患者中显示出更好的中位PFS。扩增率>2.0(10.8vs5.8个月,分别为;p=0.049)。安全性与以前的T-DM1研究一致。结论:T-DM1联合ET可显著改善HER2阳性和HR阳性MBC患者的PFS和OS。我们的研究表明,先前的帕妥珠单抗治疗加曲妥珠单抗治疗可能会降低T-DM1疗效。
    Background and Objectives: Patients with human epidermal growth factor receptor 2 (HER2) -positive, hormone receptor-positive (HR-positive) metastatic breast cancer (MBC) usually undergo trastuzumab emtansine (T-DM1) therapy in subsequent lines. Combining endocrine therapy (ET) with T-DM1 can improve treatment outcomes in this subtype. Therefore, this study aimed to investigate the benefits of using T-DM1 with ET in HER2-positive and HR-positive MBC. This study was the first to investigate the benefits of combining ET with T-DM1. Material and Methods: This study analyzed the medical records of patients with HER2-positive and HR-positive MBC who were treated with T-DM1 from June 2010 to December 2021. The patients were divided into groups based on whether they received concomitant ET with T-DM1. The primary endpoint was to determine the progression-free survival (PFS), while the secondary endpoints were overall survival (OS), objective response rate, and safety of the treatment. Results: Our analysis examined 88 patients, of whom 32 (36.4%) were treated with T-DM1 in combination with ET. The combination therapy showed a significant improvement in median PFS (15.4 vs. 6.4 months; p = 0.00004) and median OS (35.0 vs. 23.1 months; p = 0.026) compared to T-DM1 alone. The ORR was also higher in the combination group (65.6% vs. 29.3%; p = 0.026). Patients treated with pertuzumab priorly had reduced median PFS on T-DM1 compared to those who were not treated with pertuzumab (11.7 vs. 5.4 months, respectively; p < 0.01). T-DM1 demonstrated better median PFS in HER2 3+ patients compared to HER2 2+ patients, with an amplification ratio of >2.0 (10.8 vs 5.8 months, respectively; p = 0.049). The safety profiles were consistent with previous T-DM1 studies. Conclusions: The combination of T-DM1 with ET can significantly improve PFS and OS in patients with HER2-positive and HR-positive MBC. Our study suggests that prior pertuzumab treatment plus trastuzumab treatment might decrease T-DM1 efficacy.
    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
    具有延长循环的超分子递送系统,多样化功能化的潜力,很少有毒素相关的局限性被广泛研究。对于目前的研究,我们构建了一个与抗HER-2抗体曲妥珠单抗连接的线性聚甘油壳聚合物体.然后,我们通过动态二硫键将抗癌药物DM1共价加载到聚合物体中。所得曲妥珠单抗-聚合物体-DM1(Tra-PS-DM1)表现出95.3nm的平均尺寸和99.3%的显著药物装载效率%。除了其优越的稳固性,我们观察到DM1在还原条件下以受控方式快速释放。与天然聚合物囊泡相比,在HER-2阳性癌细胞中,Tra-PS-DMl已显示出极大改善的细胞摄取并显著降低IC50高达17倍。此外,Tra-PS-DM1对HER-2阳性肿瘤的生长具有极好的抑制作用;特别是,BT474肿瘤在治疗12小时后收缩至62%。具有出色的稳定性和可瞄准性,带有PG壳的Tra-PS-DM1似乎是HER-2阳性肿瘤治疗的一种有吸引力的方法.
    Supramolecular delivery systems with the prolonged circulation, the potential for diverse functionalization, and few toxin-related limitations have been extensively studied. For the present study, we constructed a linear polyglycerol-shelled polymersome attached with the anti-HER-2-antibody trastuzumab. We then covalently loaded the anticancer drug DM1 in the polymersome via dynamic disulfide bonding. The resulted trastuzumab-polymersome-DM1 (Tra-PS-DM1) exhibits a mean size of 95.3 nm and remarkable drug loading efficiency % of 99.3%. In addition to its superior stability, we observed the rapid release of DM1 in a controlled manner under reductive conditions. Compared to the native polymersomes, Tra-PS-DM1 has shown greatly improved cellular uptake and significantly reduced IC50 up to 17-fold among HER-2-positive cancer cells. Moreover, Tra-PS-DM1 demonstrated superb growth inhibition of HER-2-positive tumoroids; specifically, BT474 tumoroids shrunk up to 62% after 12 h treatment. With exceptional stability and targetability, the PG-shelled Tra-PS-DM1 appears as an attractive approach for HER-2-positive tumor treatment.
    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)是具有选择性地将其有效载荷递送至癌细胞的能力的抗癌剂。抗体-药物缀合物由通过接头连接到细胞毒性有效载荷的单克隆抗体主链组成。抗体-药物偶联效应通过膜抗原直接靶向癌细胞或通过“旁观者效应”发生。“抗体-药物缀合物已经证明对各种类型的肿瘤有效,包括乳腺癌.阿多-曲妥珠单抗emtansine是目前唯一被批准用于早期乳腺癌治疗的ADC,虽然现在有几种ADC被批准用于转移性乳腺癌。由于几个ADC在晚期乳腺癌背景下报道的变革性影响,研究人员现在在早期环境中测试更多这样的化合物,通过高效抗癌药物预示患者的益处。正在进行的试验有可能在不久的将来改变早期乳腺癌的治疗方案。这些试验旨在评估不同的治疗调制方法,根据乳腺癌复发的风险,包括治疗降级。在正在进行的临床试验中,努力确定受益最大的患者,用新型ADC追求精准医学的范式。本文综述了ADC在早期乳腺癌中的潜在作用。概述其开发的最新进展以及如何在正在进行的临床试验中实施。
    Antibody-drug conjugates (ADCs) are anticancer agents with the capacity to selectively deliver their payloads to cancer cells. Antibody-drug conjugates consist of a monoclonal antibody backbone connected by a linker to cytotoxic payloads. Antibody-drug conjugate effect occurs either by directly targeting cancer cells via membrane antigen or through \"bystander effect.\" Antibody-drug conjugates have demonstrated efficacy against various types of tumors, including breast cancer. Ado-trastuzumab emtansine is presently the only approved ADC for the treatment of breast cancer in the early setting, while several ADCs are now approved for metastatic breast cancer. Due to the transformative impact that several ADCs have reported in the setting of advanced breast cancer, researchers are now testing more of such compounds in the early setting, to portend benefits to patients through highly potent anticancer drugs. Ongoing trials hold the potential to transform treatment protocols for early breast cancer in the near future. These trials are aiming at evaluating different treatment modulation approaches, as informed by breast cancer risk of recurrence, including toward treatment de-escalation. Efforts are provided in ongoing clinical trials to identify the patients who will benefit most, to pursue paradigms of precision medicine with the novel ADCs. This review focuses on the potential role of ADCs in early breast cancer, providing an overview of the latest progress in their development and how they are implemented in ongoing clinical trials.
    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
    最近出现的抗体-药物缀合物(ADC)的显著特征包括通过单克隆抗体(mAb)和高的药物与抗体比率(DAR)靶向人表皮生长因子受体2(HER2)。Kadcyla®(T-DM1)和Enhertu®(T-Dxd)的成果证明了靶向HER2的抗体,如曲妥珠单抗,在发展的效率和价格方面表现出竞争力。此外,随着T-Dxd和Trodelvy®的到来,高清(7-8)ADC,与以前被认为是常规的中度DAR(3-4)ADC不同,他们的价值被承认。遵循这一药物发展趋势,我们努力开发一种高DARADC,使用一种直接的方法,涉及利用DM1,一种高效物质,与广泛认可的曲妥珠单抗组合。为了实现高DAR,通过具有不同长度的各种二马来酰亚胺接头的简单设计和合成,DM1与还原的半胱氨酸缀合。使用LC和MS分析,我们已经证明了我们的合成方法简单有效,产生基于曲妥珠单抗的ADC,表现出显著的均匀性。这些ADC已被实验证实在体外对癌细胞发挥抑制作用,从而肯定它们的价值是对ADC领域的值得注意的补充。
    The notable characteristics of recently emerged Antibody-Drug Conjugates (ADCs) encompass the targeting of Human Epidermal growth factor Receptor 2 (HER2) through monoclonal antibodies (mAbs) and a high ratio of drug to antibody (DAR). The achievements of Kadcyla® (T-DM1) and Enhertu® (T-Dxd) have demonstrated that HER2-targeting antibodies, such as trastuzumab, have shown to be competitive in terms of efficacy and price for development. Furthermore, with the arrival of T-Dxd and Trodelvy®, high-DAR (7-8) ADCs, which differ from the moderate DAR (3-4) ADCs that were formerly regarded as conventional, are being acknowledged for their worth. Following this trend of drug development, we endeavored to develop a high-DAR ADC using a straightforward approach involving the utilization of DM1, a highly potent substance, in combination with the widely recognized trastuzumab. To achieve a high DAR, DM1 was conjugated to reduced cysteine through the simple design and synthesis of various dimaleimide linkers with differing lengths. Using LC and MS analysis, we have demonstrated that our synthesis methodology is uncomplicated and efficacious, yielding trastuzumab-based ADCs that exhibit a remarkable degree of uniformity. These ADCs have been experimentally substantiated to exert an inhibitory effect on cancer cells in vitro, thus affirming their value as noteworthy additions to the realm of ADCs.
    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:在大多数晚期人类表皮生长因子受体2阳性(HER2+)乳腺癌患者中,抗HER2治疗由于获得性耐药性的发展而失败,可能通过磷酸肌醇-3-激酶(PI3K)信号介导。我们调查了添加taselisib,一种α选择性强效口服PI3K抑制剂,以不同的HER2为导向的方案,以改善疾病控制。
    方法:将患有晚期HER2+乳腺癌的患者(n=68)纳入该开放标签,剂量递增Ib期研究。主要终点是定义各种含有taselisib的组合的最大耐受剂量(MTD)。次要终点是安全性。探索性终点包括循环肿瘤DNA分析。该研究包括四个队列:(A)taselisib+曲妥珠单抗emtansine(T-DM1),(C)他赛利布+曲妥珠单抗和帕妥珠单抗(TP),(D)taselisib+TP+紫杉醇,和(E)taselisib+TP+氟维司群。
    结果:剂量递增后,taselisibMTD定义为每天一次4mg.治疗与显著的毒性有关,由于68例患者中有34例经历了归因于taselisib的≥3级不良事件(AE),最常见的全等级不良事件是腹泻,疲劳,和口腔粘膜炎.中位随访时间为43.8个月,队列A中MTD治疗人群的中位无进展生存期(PFS),C,E为6.3个月[95%置信区间(CI)3.2-不适用(NA)],1.7(95%CI1.4-NA)个月,和10.6个月(95%可信区间8.3-NA),分别。先前使用过T-DM1的队列A患者的中位PFS为10.4个月(95%CI2.7-NA)。
    结论:Taselisib联合HER2靶向治疗的PIK3CA靶向与有希望的疗效和实质性毒性相关。
    BACKGROUND: In most patients with advanced human epidermal growth factor receptor-2-positive (HER2+) breast cancer, anti-HER2 therapies fail due to the development of acquired resistance, potentially mediated through phosphoinositide-3-kinase (PI3K) signaling. We investigated adding taselisib, an α-selective potent oral inhibitor of PI3K, to different HER2-directed regimens in order to improve disease control.
    METHODS: Patients (n = 68) with advanced HER2+ breast cancer were enrolled to this open-label, dose-escalation phase Ib study. The primary endpoint was defining the maximal tolerated dose (MTD) for the various taselisib-containing combinations. The secondary endpoint was safety. Exploratory endpoints included circulating tumor DNA analysis. The study included four cohorts: (A) taselisib + trastuzumab emtansine (T-DM1), (C) taselisib + trastuzumab and pertuzumab (TP), (D) taselisib + TP + paclitaxel, and (E) taselisib + TP + fulvestrant.
    RESULTS: Following dose escalation, the taselisib MTD was defined as 4 mg once daily. Treatment was associated with significant toxicities, as 34 out of 68 patients experienced grade ≥3 adverse events (AEs) attributed to taselisib, the most common all-grade AEs being diarrhea, fatigue, and oral mucositis. At a median follow-up of 43.8 months, median progression-free survival (PFS) for the MTD-treated population in cohorts A, C, and E was 6.3 [95% confidence interval (CI) 3.2-not applicable (NA)] months, 1.7 (95% CI 1.4-NA) months, and 10.6 (95% CI 8.3-NA) months, respectively. The median PFS for patients in cohort A with prior T-DM1 use was 10.4 (95% CI 2.7-NA) months.
    CONCLUSIONS: PIK3CA targeting with taselisib in combination with HER2-targeted therapies was associated with both promising efficacy and substantial toxicities.
    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
    UNASSIGNED: Motivation for the study. Treatment options for HER2-positive breast cancer were evaluated, focusing on the efficacy and safety of trastuzumab-emtansine (T-DM1) compared to other anti-HER2 therapies. Main findings. Trastuzumab-deruxtecan (T-DXd) and PyroCap emerged as promising alternatives, showing substantial improvements in progression-free survival for locally advanced or metastatic breast cancer. T-DM1 showed superior efficacy to the other treatments. Implications. Our findings could inform healthcare decision-making processes to optimize strategies for HER2-positive breast cancer, and potentially improve health outcomes and quality of life. We aimed to study the efficacy and safety of trastuzumab-emtansine (T-DM1) versus other anti-HER2 therapies in HER2+ breast cancer (BC).
    UNASSIGNED: We performed a network meta-analysis (NMA) of randomized controlled trials (RCTs). Our study focused on patients undergoing treatment for unresectable locally advanced breast cancer (LABC) or metastatic breast cancer (mBC), which included regimens involving trastuzumab and taxanes. Additionally, we considered cases within the first 6 months of treatment for HER2+ early breast cancer (EBC).
    UNASSIGNED: A total of 23 RCTs and 41 reports were included in our analysis. LABC and mBC showed no statistically significant difference in any of the comparisons of T-DM1 versus the other anti-HER2+ therapies. When assessing progression-free survival (PFS), trastuzumab-deruxtecan (T-DXd) and PyroCap demonstrated greater efficacy compared to other treatments (Hazard Ratio [HR]: 3.57; 95% confidence interval [CI]: 2.75-4.63 and HR: 1.82; 95% CI: 1.35-2.44; respectively), while T-DM1 alone exhibited superior effectiveness compared to LapCap (HR: 0.65; 95% CI: 0.55-0.77), TrasCap (HR: 0.65; 95% CI: 0.46-0.91), LapCapCitu (HR: 0.60; 95% CI: 0.33-1.10), Nera (HR: 0.55; 95% CI: 0.39-0.77), and Cap (HR: 0.37; 95% CI: 0.28-0.49).
    UNASSIGNED: NMA allows a ranking based on the comparative efficacy and safety among the interventions available. Although superior to other schemes, T-DM1 showed a lower efficacy performance in PFS and overall response rate and a trend towards worse overall survival than T-DXd.
    UNASSIGNED: Motivación para realizar el estudio. Se evaluaron las opciones de tratamiento para el cáncer de mama HER-2-positivo, centrándose en la eficacia y seguridad de trastuzumab-emtansina (T-DM1) en comparación con otras terapias anti-HER-2. Principales hallazgos. Trastuzumab-deruxtecan (T-DXd)y PyroCap surgieron como alternativas prometedoras, mostrando mejoras sustanciales en la sobrevida libre de progresión para el cáncer de mama localmente avanzado o metastásico. T-DM1 mostró una eficacia superior a la de los demás tratamientos. Implicancias. Nuestros hallazgos podrían informar los procesos de toma de decisiones sanitarias para optimizar las estrategias para el cáncer de mama HER-2-positivo, y potencialmente mejorar los resultados de salud y la calidad de vida. Nuestro objetivo fue estudiar la eficacia y la seguridad de trastuzumab-emtansina (T-DM1) en comparación con otras terapias anti-HER-2 en el cáncer de mama (CM) HER-2 positivo.
    UNASSIGNED: Realizamos un metaanálisis de red (NMA, por sus siglas en inglés) de ensayos clínicos aleatorizados (ECA). Nuestro estudio se centró en pacientes sometidos al tratamiento para el cáncer de mama localmente avanzado no resecable (CMLA) o cáncer de mama metastásico (CMm), que incluía esquemas con trastuzumab y taxanos. Además, consideramos casos dentro de los primeros 6 meses de tratamiento para el cáncer de mama temprano (CMT) HER-2 positivo.
    UNASSIGNED: Se incluyeron en nuestro análisis un total de 23 ECA y 41 reportes. En CMLA y CMm, no se observaron diferencias estadísticamente significativas en ninguna de las comparaciones entre T-DM1 y otras terapias anti-HER-2 positivo. Al evaluar la sobrevida libre de progresión (SLP), trastuzumab-deruxtecan (T-DXd) y PyroCap demostraron una mayor eficacia en comparación con otros tratamientos (Hazard Ratio [HR]: 3,57; intervalo de confianza al 95% [IC 95%]: 2,75-4,63 y HR: 1.82; IC 95%: 1,35-2,44; respectivamente), mientras que T-DM1 por sí solo mostró una efectividad superior en comparación con LapCap (HR: 0,65; IC 95%: 0,55-0,77), TrasCap (HR: 0,65; IC 95%: 0,46-0,91), LapCapCitu (HR: 0,60; IC 95%: 0,33-1,1), Nera (HR: 0,55; IC 95%: 0,39-0,77) y Cap (HR: 0,37; IC 95%: 0,28-0,49).
    UNASSIGNED: Este NMA estableció un ranking basado en la eficacia y seguridad comparativas entre las intervenciones disponibles. Aunque superior a otros esquemas, T-DM1 mostró una menor eficacia en la SLP y la tasa de respuesta objetiva, y una tendencia hacia una sobrevida global peor que T-DXd.
    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:DESTINY-Breast03是随机的,多中心,开放标签,在人类表皮生长因子受体2(HER2)阳性转移性乳腺癌(mBC)患者中,曲妥珠单抗地克替康(T-DXd)与曲妥珠单抗(T-DM1)的III期研究,患者既往接受曲妥珠单抗和紫杉烷治疗.在主要分析中报告了无进展生存期(PFS)相对于T-DM1的统计学显着改善。这里,我们报告了基线时有无脑转移(BMs)患者的探索性疗效数据.
    方法:患者被随机分配1:1接受T-DXd5.4mg/kg或T-DM13.6mg/kg。临床上不活跃/无症状的患者符合资格。根据改良的RECIST测量病变,1.1版。结果包括盲化独立中央审查(BICR)的PFS,客观反应率(ORR),和颅内ORR根据BICR。
    结果:截至2021年5月21日,43/261例随机接受T-DXd治疗的患者和39/263例随机接受T-DM1治疗的患者在基线时具有BMs,根据研究者的评估。在具有基线BMs的患者中,T-DXd臂中的20/43和T-DM1臂中的19/39未接受局部BM治疗。对于有BMS的患者,T-DXd的中位PFS为15.0个月[95%置信区间(CI)12.5-22.2个月],T-DM1为3.0个月(95%CI2.8-5.8个月);风险比(HR)0.25(95%CI0.13-0.45).对于没有BMS的患者,T-DXd与T-DM1的7.1个月(95%CI为5.6~9.7个月)相比,中位PFS未达到(95%CI为22.4个月-不可估计);HR0.30(95%CI为0.22~0.40).对于有和没有BMs的患者,T-DXd的全身ORR为67.4%,T-DM1为20.5%,T-DXd为82.1%,T-DM1为36.6%。分别。T-DXd的颅内ORR为65.7%,T-DM1为34.3%。
    结论:与T-DM1相比,接受曲妥珠单抗和紫杉烷治疗后病情进展的HER2阳性mBC患者从T-DXd治疗中获得了实质性益处,包括那些基线BMs。
    BACKGROUND: DESTINY-Breast03 is a randomized, multicenter, open-label, phase III study of trastuzumab deruxtecan (T-DXd) versus trastuzumab emtansine (T-DM1) in patients with human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer (mBC) previously treated with trastuzumab and a taxane. A statistically significant improvement in progression-free survival (PFS) versus T-DM1 was reported in the primary analysis. Here, we report exploratory efficacy data in patients with and without brain metastases (BMs) at baseline.
    METHODS: Patients were randomly assigned 1 : 1 to receive T-DXd 5.4 mg/kg or T-DM1 3.6 mg/kg. Patients with clinically inactive/asymptomatic BMs were eligible. Lesions were measured as per modified RECIST, version 1.1. Outcomes included PFS by blinded independent central review (BICR), objective response rate (ORR), and intracranial ORR as per BICR.
    RESULTS: As of 21 May 2021, 43/261 patients randomized to T-DXd and 39/263 patients randomized to T-DM1 had BMs at baseline, as per investigator assessment. Among patients with baseline BMs, 20/43 in the T-DXd arm and 19/39 in the T-DM1 arm had not received prior local BM treatment. For patients with BMs, median PFS was 15.0 months [95% confidence interval (CI) 12.5-22.2 months] for T-DXd versus 3.0 months (95% CI 2.8-5.8 months) for T-DM1; hazard ratio (HR) 0.25 (95% CI 0.13-0.45). For patients without BMs, median PFS was not reached (95% CI 22.4 months-not estimable) for T-DXd versus 7.1 months (95% CI 5.6-9.7 months) for T-DM1; HR 0.30 (95% CI 0.22-0.40). Confirmed systemic ORR was 67.4% for T-DXd versus 20.5% for T-DM1 and 82.1% for T-DXd versus 36.6% for T-DM1 for patients with and without BMs, respectively. Intracranial ORR was 65.7% with T-DXd versus 34.3% with T-DM1.
    CONCLUSIONS: Patients with HER2-positive mBC whose disease progressed after trastuzumab and a taxane achieved a substantial benefit from treatment with T-DXd compared with T-DM1, including those with baseline BMs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号