pertuzumab

帕妥珠单抗
  • 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
    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)

  • DOI:
    文章类型: Journal Article
    HER2受体酪氨酸激酶是表皮生长因子受体家族成员之一,包括EGFR,HER3和HER4已知它们在正常发育和癌症中起关键作用。乳腺癌的一个子集与HER2基因有关,在20-25%的浸润性乳腺癌中扩增和/或过表达,并与肿瘤对化疗的耐药性相关,转移性乳腺癌(MBC)和患者生存率低。受体酪氨酸激酶抑制剂的出现改善了HER2阳性乳腺癌的预后;然而,HER2+MBC总是进展(获得性抗性或从头抗性)。基于单克隆抗体的药物(大分子TKIs)靶向HER2的细胞外结合结构域;而小分子TKIs在细胞内作用以抑制增殖和存活信号。我们回顾了TKIs的作用方式,以期显示哪些TKIs可以结合在纳米颗粒中,以受益于纳米技术的力量(降低毒性,改善疏水性药物的溶解度,循环半衰期长,绕过外排泵并防止网状内皮系统(单核吞噬细胞系统)捕获。纳米治疗剂还介导掺入纳米颗粒中的多种药物的药代动力学和生物分布的同步。提到了目前正在研究的有或没有纳米颗粒递送的新型TKIs,并提出了基于纳米的策略来改善其交付。目前在临床实践中的免疫疗法,讨论了临床试验或临床前阶段。然而,免疫疗法仅在相对较小的患者亚群中有效。将纳米医学与免疫疗法相结合可以提高治疗效果,通过将“冷”非免疫反应性肿瘤和转移转化为“热”免疫反应性病变。
    The HER2 receptor tyrosine kinase is a member of the epidermal growth factor receptor family which includes EGFR, HER3 and HER4. They are known to play critical roles in both normal development and cancer. A subset of breast cancers is associated with the HER2 gene, which is amplified and/or overexpressed in 20-25% of invasive breast cancers and is correlated with tumor resistance to chemotherapy, Metastatic Breast Cancer (MBC) and poor patient survival. The advent of receptor tyrosine kinase inhibitors has improved the prognosis of HER2-postive breast cancers; however, HER2+MBC invariably progresses (acquired resistance or de novo resistance). The monoclonal antibody-based drugs (large molecule TKIs) target the extracellular binding domain of HER2; while the small molecule TKIs act intracellularly to inhibit proliferation and survival signals. We reviewed the modes of action of the TKIs with a view to showing which of the TKIs could be combined in nanoparticles to benefit from the power of nanotechnology (reduced toxicity, improved solubility of hydrophobic drugs, long circulation half-lives, circumventing efflux pumps and preventing capture by the reticuloendothelial system (mononuclear phagocyte system). Nanotherapeutics also mediate the synchronization of the pharmacokinetics and biodistribution of multiple drugs incorporated in the nanoparticles. Novel TKIs that are currently under investigation with or without nanoparticle delivery are mentioned, and nano-based strategies to improve their delivery are suggested. Immunotherapies currently in clinical practice, clinical trials or at the preclinical stage are discussed. However, immunotherapy only works well in relatively small subsets of patients. Combining nanomedicine with immunotherapy can boost therapeutic outcomes, by turning \"cold\" non-immunoresponsive tumors and metastases into \"hot\" immunoresponsive lesions.
    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
    我们的目的是评估曲妥珠单抗emtansine在先前接受帕妥珠单抗联合曲妥珠单抗和紫杉烷治疗的转移性乳腺癌患者中的疗效和安全性。我们回顾了被诊断为人类表皮生长因子受体2(HER-2)阳性转移性乳腺癌并在2014年1月至2021年1月期间从25个癌症中心接受帕妥珠单抗和TDM-1的患者的医疗记录。Kaplan-Meier方法估计无进展生存期(PFS)和总生存期(OS)。此外,客观反应率(ORR),临床获益率(CBR),并对安全性进行了评估。一百五十三名患者被包括在内,79.1%的患者在二线接受TDM-1,90.8%有内脏转移,30.7%有中枢神经系统受累。TDM-1的PFS和OS根据先前行(第2行或2行以上)转移部位(内脏和非内脏)的数量和中枢神经转移的存在进行评估。在TDM-1治疗中,二线治疗的PFS为10个月(95%CI:7.7-12.2);这在统计学上高于后期的PFS,这是六个月(95%CI:3.3至8.6)(p=0.004)。二线接受TDM-1治疗的患者的中位OS时间为25个月(95%CI:21.0至28.9),二线接受时间晚于二线的患者的中位OS时间为19个月(95%CI:12.3至25.6)(p=0.175)。有和没有内脏和中枢神经转移的患者的PFS时间没有显着差异。我们的研究表明,TDM-1对使用帕妥珠单抗的患者也有效,与在后一行中使用相比,在第二行中使用时对PFS的贡献很大,并且在操作系统中没有任何区别。
    We aimed to evaluate the efficacy and safety of trastuzumab emtansine in patients with metastatic breast cancer previously treated with pertuzumab plus trastuzumab and taxane. We reviewed the medical records of patients who were diagnosed with Human Epidermal Growth Factor Receptor 2 (HER-2) positive metastatic breast cancer and received pertuzumab and then TDM-1 between January 2014 and January 2021 from twenty- five cancer centers. The Kaplan- Meier method estimated progression-free survival (PFS) and overall survival (OS). Additionally, objective response rate (ORR), clinical benefit rate (CBR), and safety were evaluated. One hundred fifty-three patients were included,79.1% of the patients received TDM-1 in the second line, 90.8% had visceral metastasis, and 30.7% had central nervous system involvement. The PFS and OS of TDM-1 were evaluated according to the number of previous lines (on the 2nd line or more than two lines) metastatic sites (visceral and non-visceral) and the presence of central nervous metastasis. In TDM-1 therapy, PFS in second line therapy was ten months (95% CI: 7.7 - 12.2); this was statistically higher than later-line PFS, which was six months (95% CI: 3.3 to 8.6) (p = 0.004). The median OS time was 25 months (95% CI: 21.0 to 28.9) in patients treated with TDM-1 in the second line and 19 months (95% CI: 12.3 to 25.6) in patients who received later than the second line(p = 0.175). There were no significant differences in PFS time of patients with and without visceral and central nervous metastases. Our study showed that TDM-1 was also effective in patients using pertuzumab, contributes significantly to PFS when used in the second line compared to its use in the later line, and does not make any difference in OS.
    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:在曲妥珠单抗(H)和标准化疗(CT)基础上增加帕妥珠单抗(P)作为HER2+乳腺癌(BC)患者的新辅助治疗(NaT),已显示增加病理完全缓解(pCR)率,没有主要的安全问题。NeoPowER试验的目的是评估真实世界人群中P+H+CT的安全性和有效性。
    方法:我们回顾性回顾了II-III期的医疗记录,接受NaT治疗的HER2BC患者:在5个EmiliaRomagna机构中接受PHCT(新功率组)的患者与接受HCT的历史组(对照组)进行了比较。主要终点是安全性,次要终点是pCR率,DRFS和OS及其与NaT和其他潜在变量的相关性。
    结果:纳入260例患者,48%接受P+H+CT,其中44%的人作为CT的一部分接受了蒽醌治疗,与对照组的83%相比。毒性特征相似,排除新电源组更频繁的腹泻(20%vs.9%)。三例患者左心室射血分数(LVEF)显着降低,都接受蒽环类药物治疗.pCR率为46%(P+H+CT)和40%(H+CT)(p=0.39)。P的添加仅在接受无去甲方案的患者中与pCR具有统计学相关性(OR=3.05,p=0.047)。术前蒽环类药物的使用(OR=1.81,p=0.03)和NaT的持续时间(OR=1.18,p=0.02)与pCR有统计学关系。对照组发生12/21的远处复发事件和14/17的死亡。达到pCR的患者DRFS显着增加(HR=0.23,p=0.009)。
    结论:在H和CT中添加新辅助P是安全的。除了腹泻,2级>2级的不良事件发生率两组间无差异.当添加到H+CT时,P没有增加心脏毒性,尽管如此,在我们人群中,所有心脏事件均发生在接受蒽环类药物治疗的患者中.没有统计学意义,在接受新辅助P+H+CT的患者中,可以实现更高的pCR率.该研究未显示P的增加与长期结果之间的统计学显着相关性。
    BACKGROUND: The addition of pertuzumab (P) to trastuzumab (H) and standard chemotherapy (CT) as neoadjuvant treatment (NaT) for patients with HER2 + breast cancer (BC), has shown to increase the pathological complete response (pCR) rate, without main safety concerns. The aim of NeoPowER trial is to evaluate safety and efficacy of P + H + CT in a real-world population.
    METHODS: We retrospectively reviewed the medical records of stage II-III, HER2 + BC patients treated with NaT: who received P + H + CT (neopower group) in 5 Emilia Romagna institutions were compared with an historical group who received H + CT (control group). The primary endpoint was the safety, secondary endpoints were pCR rate, DRFS and OS and their correlation to NaT and other potential variables.
    RESULTS: 260 patients were included, 48% received P + H + CT, of whom 44% was given anthraciclynes as part of CT, compared to 83% in the control group. The toxicity profile was similar, excluding diarrhea more frequent in the neopower group (20% vs. 9%). Three patients experienced significant reductions in left ventricular ejection fraction (LVEF), all receiving anthracyclines. The pCR rate was 46% (P + H + CT) and 40% (H + CT) (p = 0.39). The addition of P had statistically correlation with pCR only in the patients receiving anthra-free regimens (OR = 3.05,p = 0.047). Preoperative use of anthracyclines (OR = 1.81,p = 0.03) and duration of NaT (OR = 1.18,p = 0.02) were statistically related to pCR. 12/21 distant-relapse events and 14/17 deaths occurred in the control group. Patients who achieve pCR had a significant increase in DRFS (HR = 0.23,p = 0.009).
    CONCLUSIONS: Adding neoadjuvant P to H and CT is safe. With the exception of diarrhea, rate of adverse events of grade > 2 did not differ between the two groups. P did not increase the cardiotoxicity when added to H + CT, nevertheless in our population all cardiac events occurred in patients who received anthracycline-containing regimens. Not statistically significant, higher pCR rate is achievable in patients receiving neoadjuvant P + H + CT. The study did not show a statistically significant correlation between the addition of P and long-term outcomes.
    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
    在过去的几年里,曲妥珠单抗和紫杉醇联合治疗已成为人类表皮生长因子受体2(HER2)阳性乳腺癌的重要选择.小分子酪氨酸激酶抑制剂(TKIs)可以为HER2阳性乳腺癌患者带来临床益处。然而,这两种方案的疗效和安全性尚未进行比较.本研究探讨了吡唑替尼联合曲妥珠单抗和白蛋白结合型紫杉醇(nab-pacytaxel)的疗效和安全性。
    纳入2020年11月至2022年6月在牡丹江市肿瘤医院治疗的新诊断HER2阳性早期或局部晚期乳腺癌患者。对照组接受帕妥珠单抗联合nab-紫杉醇,而吡唑替尼组接受吡唑替尼联合帕妥珠单抗和nab-紫杉醇作为治疗,在3周的4个周期的周期。这项研究的主要终点是总病理完全缓解(tpCR)率,乳腺病理完全缓解(bpCR)率,次要终点包括无进展生存期(PFS),客观反应率(ORR),和不良事件(AE)的发生。
    共有72名患者被纳入研究并完成研究治疗。基线特征在这两个臂之间很好地平衡。在对照组中,tPCR率为23.68%,bpCR率为47.36%。在pyrotinib组中,tPCR率为47.06%,bpCR率为64.71%。吡罗替尼组tPCR率显著高于对照组(P=0.049)。吡唑替尼组的ORR(67.65%)明显高于对照组(42.11%,P=0.04)。对照组的中位PFS(mPFS)为9.24个月,平均PFS为10.01±0.44个月[95%置信区间(CI):9.14-10.88个月]。在pyrotinib组中,mPFS为9.74个月,平均PFS为11.25±0.29个月(95%CI:10.67-11.82个月)。吡唑替尼组PFS明显长于对照组(P=0.045)。安全性结果显示,对照组不良事件的总发生率为68.42%,3级不良反应率为21.05%。在pyrotinib组中,AEs的总发生率为79.41%,3级不良反应率为29.41%。两组比较差异无统计学意义(P>0.05)。
    吡罗替尼组在新辅助治疗HER2阳性乳腺癌的近期疗效优于对照组。这种治疗方案可以延长PFS1年,用药期间的安全性是可控的。本研究还存在一定的局限性,样本量相对较小,随访时间相对较短,进一步大规模,多中心,需要进行随机对照试验来验证这种双靶点治疗方案的临床价值.
    UNASSIGNED: In the past few years, the combination of trastuzumab and paclitaxel has become an important option for human epidermal growth factor receptor-2 (HER2)-positive breast cancer. Small molecule tyrosine kinase inhibitors (TKIs) can bring clinical benefit to HER2-positive breast cancer patients. However, the efficacy and safety of these two regimens have not been compared. This study explored the efficacy and safety of pyrotinib combined with trastuzumab and albumin-bound paclitaxel (nab-paclitaxel).
    UNASSIGNED: Patients with newly diagnosed HER2-positive early or locally advanced breast cancer treated at The Tumor Hospital of Mudanjiang City from November 2020 to June 2022 were included. The control group received pertuzumab in combination with nab-paclitaxel, whereas the pyrotinib group received pyrotinib in combination with pertuzumab and nab-paclitaxel as treatment, in a 3-week cycle for 4 cycles. The primary endpoints of this study were total pathological complete response (tpCR) rate, breast pathological complete response (bpCR) rate, and the secondary endpoints included progression-free survival (PFS), objective response rate (ORR), and the occurrence of adverse events (AEs).
    UNASSIGNED: A total of 72 patients were enrolled in the study and completed the study treatment. Baseline characteristics were well balanced between these two arms. In the control group, the tPCR rate was 23.68%, and the bpCR rate was 47.36%. In the pyrotinib group, the tPCR rate was 47.06%, and the bpCR rate was 64.71%. The tPCR rate in the pyrotinib group was significantly higher than that in the control group (P=0.049). The ORR in the pyrotinib group (67.65%) was significantly higher than that in the control group (42.11%, P=0.04 ). The median PFS (mPFS) for the control group was 9.24 months, with a mean PFS of 10.01±0.44 months [95% confidence interval (CI): 9.14-10.88 months]. In the pyrotinib group, mPFS was 9.74 months, with a mean PFS of 11.25±0.29 months (95% CI: 10.67-11.82 months). The PFS in the pyrotinib group was significantly longer than that in the control group (P=0.045). Safety results showed that the overall incidence of AEs in the control group was 68.42%, with a 3-grade adverse reaction rate of 21.05%. In the pyrotinib group, the overall incidence of AEs was 79.41%, with a 3-grade adverse reaction rate of 29.41%. The difference between the two groups was not statistically significant (P>0.05).
    UNASSIGNED: Pyrotinib group in neoadjuvant treatment for HER2 positive breast cancer has obvious short-term efficacy advantages over control group. This treatment regimen can prolong PFS for 1 year, and the safety during medication is controllable. This study still has some limitations, with the relatively small sample size and relatively short follow-up period, and a further large-scale, multicenter, randomized controlled trial is necessary to verify the clinical value of this dual-target treatment regimen.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Clinical Trial, Phase III
    背景:HER2阳性,雌激素受体阳性乳腺癌(HER2+,ER+BC)是一种独特的疾病亚型,与HER2+相比,对化疗加HER2靶向治疗的反应较差,ER阴性BC。双向串扰导致HER2和ER途径的合作,这可能会驱动治疗阻力;因此,同时联合靶向可以优化HER2+患者的治疗效果和生存结果,ER+BC。HER2+转移性BC(mBC)患者的一线(1L)治疗是帕妥珠单抗,曲妥珠单抗,紫杉烷化疗.在临床实践中,双重HER2阻断加上固定数量的化疗周期作为诱导治疗,以最大化肿瘤反应,随后给予HER2靶向维持治疗作为长期疾病控制的更耐受方案。对于肿瘤共表达ER的患者,可以增加维持内分泌治疗(ET),但由于缺乏来自随机临床试验的数据,研究维持ET+双重HER2阻断与单独使用双重HER2阻断相比的优越性,因此摄取量存在差异.Giredestrant,一种新型口服选择性ER拮抗剂和降解剂,在ER+患者的I-II期试验中显示出有希望的临床活性和可管理的安全性,HER2阴性BC,在HER2共表达的患者中具有治疗潜力。
    方法:第三阶段,随机化,开放标签,双臂研究旨在招募812名HER2+患者,ER+局部晚期(LA)/mBC进入诱导期(帕妥珠单抗和曲妥珠单抗的固定剂量组合用于皮下注射[PHFDCSC]加紫杉烷),使730名患者1:1随机分配至维持期(giredestrant加PHFDCSC或PHFDCSC[加可选ET]),按疾病部位分层(内脏与非内脏),LA/转移性表现的类型(从头与复发),对诱导治疗的最佳总体反应(部分/完全反应与稳定疾病),并打算给ET(是与否)。主要终点是研究者评估的无进展生存期。次要终点包括总生存期,客观反应率,临床获益率,响应的持续时间,安全,和患者报告的结果。
    结论:heredERABC将讨论giredeststrant加双重HER2阻断是否优于单独的双重HER2阻断,为了告知在临床实践中使用该组合来维持HER2患者的1L治疗,ER+LA/mBC。
    背景:ClinicalTrials.gov,NCT05296798;2022年3月25日注册。协议3.0版(2022年11月18日)。
    背景:F霍夫曼-拉罗氏有限公司,Grenzacherstrasse1244070,巴塞尔,瑞士。
    BACKGROUND: HER2-positive, estrogen receptor-positive breast cancer (HER2+, ER+ BC) is a distinct disease subtype associated with inferior response to chemotherapy plus HER2-targeted therapy compared with HER2+, ER-negative BC. Bi-directional crosstalk leads to cooperation of the HER2 and ER pathways that may drive treatment resistance; thus, simultaneous co-targeting may optimize treatment impact and survival outcomes in patients with HER2+, ER+ BC. First-line (1L) treatment for patients with HER2+ metastatic BC (mBC) is pertuzumab, trastuzumab, and taxane chemotherapy. In clinical practice, dual HER2 blockade plus a fixed number of chemotherapy cycles are given as induction therapy to maximize tumor response, with subsequent HER2-targeted maintenance treatment given as a more tolerable regimen for long-term disease control. For patients whose tumors co-express ER, maintenance endocrine therapy (ET) can be added, but uptake varies due to lack of data from randomized clinical trials investigating the superiority of maintenance ET plus dual HER2 blockade versus dual HER2 blockade alone. Giredestrant, a novel oral selective ER antagonist and degrader, shows promising clinical activity and manageable safety across phase I-II trials of patients with ER+, HER2-negative BC, with therapeutic potential in those with HER2 co-expression.
    METHODS: This phase III, randomized, open-label, two-arm study aims to recruit 812 patients with HER2+, ER+  locally advanced (LA)/mBC into the induction phase (fixed-dose combination of pertuzumab and trastuzumab for subcutaneous injection [PH FDC SC] plus a taxane) to enable 730 patients to be randomized 1:1 to the maintenance phase (giredestrant plus PH FDC SC or PH FDC SC [plus optional ET]), stratified by disease site (visceral versus non-visceral), type of LA/metastatic presentation (de novo versus recurrent), best overall response to induction therapy (partial/complete response versus stable disease), and intent to give ET (yes versus no). The primary endpoint is investigator-assessed progression-free survival. Secondary endpoints include overall survival, objective response rate, clinical benefit rate, duration of response, safety, and patient-reported outcomes.
    CONCLUSIONS: heredERA BC will address whether giredestrant plus dual HER2 blockade is superior to dual HER2 blockade alone, to inform the use of this combination in clinical practice for maintenance 1L treatment of patients with HER2+, ER+ LA/mBC.
    BACKGROUND: ClinicalTrials.gov, NCT05296798; registered on March 25, 2022. Protocol version 3.0 (November 18, 2022).
    BACKGROUND: F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124 4070, Basel, Switzerland.
    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:与标准TCbHP方案相比,随机2期新峰研究检查了多西他赛+卡铂+曲妥珠单抗+帕妥珠单抗(T-DM1+P)后新辅助曲妥珠单抗(T-DM1+P)的有效性。我们先前报道,TCbHP和T-DM1+P在新辅助治疗后的pCR率倾向于更高。我们对术后5年的预后进行了探索性分析。
    方法:新辅助治疗TCbHP(6个周期;A组),TCbHP(4个周期),随后T-DM1+P(4个周期;B组),T-DM1+P(4个周期;C组,+2个周期的应答者)进行比较。4个周期后,C组无反应者改用基于蒽环类的方案。我们评估了5年无病生存期(DFS),远程DFS(DDFS),总生存率(OS)。
    结果:数据来自203名患者(A-C组的50、52和101,分别)进行了分析。DFS没有发现显著的组间差异,DDFS,或操作系统。5年DFS率(95%CI)为91.8%(79.6-96.8%),92.3%(80.8-97.0%),A-C组88.0%(79.9-93.0%),分别。TCbHP随后是T-DM1P和T-DM1P,并在反应指导下增加蒽环类药物治疗,其长期预后与TCbHP相似。
    结论:在T-DM1+P新辅助治疗后达到pCR的患者中,可以考虑省略蒽环类佐剂,而对于有残留疾病的非pCR患者,应调整治疗方法。具有响应指导的治疗调整的T-DM1+P可用于最小化毒性。
    UMIN-CTR,UMIN000014649,2014年7月25日预期注册。一些研究结果在ESMO乳腺癌2023年的小型口头会议上发表(柏林,德国,2023年5月11日至13日)。
    OBJECTIVE: The randomized phase 2 Neo-peaks study examined usefulness of neoadjuvant trastuzumab emtansine + pertuzumab (T-DM1 + P) following docetaxel + carboplatin + trastuzumab + pertuzumab (TCbHP) as compared with the standard TCbHP regimen. We previously reported that pCR rate after neoadjuvant therapy tended to be higher with TCbHP followed by T-DM1 + P. We conducted an exploratory analysis of prognosis 5 years after surgery.
    METHODS: Neoadjuvant treatment with TCbHP (6 cycles; group A), TCbHP (4 cycles) followed by T-DM1 + P (4 cycles; group B), and T-DM1 + P (4 cycles; group C, + 2 cycles in responders) were compared. Group C non-responders after 4 cycles were switched to an anthracycline-based regimen. We evaluated 5-year disease-free survival (DFS), distant DFS (DDFS), and overall survival (OS).
    RESULTS: Data from 203 patients (50, 52, and 101 in groups A-C, respectively) were analyzed. No significant intergroup differences were found for DFS, DDFS, or OS. The 5-year DFS rates (95% CI) were 91.8% (79.6-96.8%), 92.3% (80.8-97.0%), and 88.0% (79.9-93.0%) in groups A-C, respectively. TCbHP followed by T-DM1 + P and T-DM1 + P with response-guided addition of anthracycline therapy resulted in similar long-term prognosis to that of TCbHP.
    CONCLUSIONS: In patients who achieved pCR after neoadjuvant therapy with T-DM1 + P, omission of adjuvant anthracycline may be considered, whereas treatment should be adjusted for non-pCR patients with residual disease. T-DM1 + P with response-guided treatment adjustment may be useful for minimizing toxicity.
    UNASSIGNED: UMIN-CTR, UMIN000014649, prospectively registered July 25, 2014. Some of the study results were presented as a Mini Oral session at the ESMO Breast Cancer 2023 (Berlin, Germany, 11-13 May 2023).
    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
    人类表皮生长因子受体2阳性(HER2+)转移性乳腺癌(MBC)的一线治疗涉及曲妥珠单抗的组合,帕妥珠单抗,和紫杉烷(TPH)。这项研究评估了曲妥珠单抗和帕妥珠单抗(PH)在常规实践中的疗效。按照乌拉圭国家资源基金(FNR)的治疗方案,类似于临床试验。
    评估了2008年至2022年间根据FNR方案接受PH治疗的晚期MBC患者。Kaplan-Meyer方法和对数秩检验用于分析总生存期(OS)。人口统计学和临床变量,包括年龄,更年期状态,和激素受体(HR),进行了分析。
    该研究包括318名接受PH治疗的患者。中位年龄为56岁,63.2%绝经后,60.4%HR和HER-2阳性。中位随访时间为17.2个月,中位OS为29个月.OS根据HR状态和不同部位转移的存在而变化,大脑患者明显较低,皮肤/皮下,和肺转移。此外,与公立机构相比,在私立机构接受治疗的患者的OS更高。
    这项研究证明了乌拉圭临床试验和临床现实之间的肿瘤治疗效果的差异,强调真实环境研究对拉丁美洲更具代表性和有效药物的重要性。
    UNASSIGNED: The first-line treatment for human epidermal growth factor receptor 2-positive (HER2+) metastatic breast cancer (MBC) involves a combination of trastuzumab, pertuzumab, and a taxane (TPH). This study assessed the efficacy of trastuzumab and pertuzumab (PH) in routine practice, following the treatment protocols of Uruguay\'s National Resources Fund (FNR), akin to clinical trials.
    UNASSIGNED: Patients with advanced MBC treated with PH between 2008 and 2022 per FNR protocols were evaluated. The Kaplan-Meyer method and log-rank test were utilized for analyzing overall survival (OS). Demographic and clinical variables, including age, menopausal status, and hormone receptors (HR), were analyzed.
    UNASSIGNED: The study included 318 PH-treated patients. The median age was 56 years, with 63.2% being postmenopausal and 60.4% HR and HER-2 positive. With a median follow-up of 17.2 months, the median OS was 29 months. OS varied based on HR status and the presence of metastases at different sites, significantly lower in patients with brain, cutaneous/subcutaneous, and pulmonary metastases. Additionally, OS was higher in patients treated at private institutions compared to public ones.
    UNASSIGNED: This study demonstrates the disparity in oncological treatment efficacy between clinical trials and clinical reality in Uruguay, emphasizing the importance of authentic environment research for more representative and effective medicine in Latin America.
    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
    在人表皮生长因子受体2(HER2)阳性(+)乳腺癌患者中经常发生对曲妥珠单抗的原发性耐药,并且仍然是临床挑战。吡罗替尼是一种新型的酪氨酸激酶抑制剂,已在HER2+乳腺癌的治疗中显示出疗效。然而,吡唑替尼治疗原发性曲妥珠单抗耐药的HER2+乳腺癌的疗效尚不清楚.
    用曲妥珠单抗治疗对曲妥珠单抗敏感或主要耐药的HER2+乳腺癌细胞,pyrotinib,或组合。细胞增殖,迁移,入侵,和HER2下游信号通路进行分析。在体外乳腺癌细胞和对曲妥珠单抗具有主要抗性的异种移植小鼠模型中比较了吡唑替尼加曲妥珠单抗和帕妥珠单抗加曲妥珠单抗的效果。
    吡罗替尼通过抑制磷酸肌醇3-激酶(PI3K)/蛋白激酶B(AKT)和大鼠肉瘤病毒(RAS)/快速加速纤维肉瘤(RAF)/丝裂原活化蛋白激酶(MAPK)/细胞外信号调节激酶(ERK)途径对曲妥珠单抗敏感的HER2乳腺癌细胞具有治疗作用。在原代曲妥珠单抗耐药细胞中,吡唑替尼抑制细胞生长,迁移,入侵,和HER2下游通路,而曲妥珠单抗没有影响.与单独使用吡唑替尼相比,与曲妥珠单抗的组合没有显示出增加的效果。与帕妥珠单抗加曲妥珠单抗相比,在曲妥珠单抗耐药HER2+乳腺癌异种移植模型中,吡罗替尼联合曲妥珠单抗在抑制乳腺癌细胞中的细胞增殖和HER2下游通路以及肿瘤生长方面更有效.
    含有吡罗替尼的治疗在HER2+乳腺癌细胞中表现出抗癌作用敏感,对曲妥珠单抗具有原发性耐药性。值得注意的是,在抑制对曲妥珠单抗的原发性耐药的HER2+乳腺癌的肿瘤生长和HER2下游通路方面,比曲妥珠单抗联合曲妥珠单抗更有效.这些发现支持联合细胞内小分子和细胞外抗体的双重抗HER2治疗的治疗功效的临床测试。
    UNASSIGNED: Primary resistance to trastuzumab frequently occurs in human epidermal growth factor receptor 2 (HER2)-positive (+) breast cancer patients and remains a clinical challenge. Pyrotinib is a novel tyrosine kinase inhibitor that has shown efficacy in the treatment of HER2+ breast cancer. However, the efficacy of pyrotinib in HER2+ breast cancer with primary trastuzumab resistance is unknown.
    UNASSIGNED: HER2+ breast cancer cells sensitive or primarily resistant to trastuzumab were treated with trastuzumab, pyrotinib, or the combination. Cell proliferation, migration, invasion, and HER2 downstream signal pathways were analyzed. The effects of pyrotinib plus trastuzumab and pertuzumab plus trastuzumab were compared in breast cancer cells in vitro and a xenograft mouse model with primary resistance to trastuzumab.
    UNASSIGNED: Pyrotinib had a therapeutic effect on trastuzumab-sensitive HER2+ breast cancer cells by inhibiting phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) and rat sarcoma virus (RAS)/rapidly accelerated fibrosarcoma (RAF)/mitogen-activated protein kinase (MAPK)/extracellular-signal regulated kinase (ERK) pathways. In primary trastuzumab-resistant cells, pyrotinib inhibited cell growth, migration, invasion, and HER2 downstream pathways, whereas trastuzumab had no effects. The combination with trastuzumab did not show increased effects compared with pyrotinib alone. Compared with pertuzumab plus trastuzumab, pyrotinib plus trastuzumab was more effective in inhibiting cell proliferation and HER2 downstream pathways in breast cancer cells and tumor growth in a trastuzumab-resistant HER2+ breast cancer xenograft model.
    UNASSIGNED: Pyrotinib-containing treatments exhibited anti-cancer effects in HER2+ breast cancer cells sensitive and with primary resistance to trastuzumab. Notably, pyrotinib plus trastuzumab was more effective than trastuzumab plus pertuzumab in inhibiting tumor growth and HER2 downstream pathways in HER2+ breast cancer with primary resistance to trastuzumab. These findings support clinical testing of the therapeutic efficacy of dual anti-HER2 treatment combining an intracellular small molecule with an extracellular antibody.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号