Advances in Clinical Medicine
Vol. 13  No. 07 ( 2023 ), Article ID: 69517 , 7 pages
10.12677/ACM.2023.1371686

曲妥珠单抗靶向治疗HER2阳性乳腺癌的 研究进展

胥世霜1,2

1延安大学第一临床医学院,陕西 延安

2延安大学附属医院肿瘤科,陕西 延安

收稿日期:2023年6月25日;录用日期:2023年7月19日;发布日期:2023年7月27日

摘要

乳腺癌的发展趋势日益增长,其中人表皮生长因子受体2 (HER2)阳性乳腺癌占所有乳腺癌的15%~20%。HER2阳性乳腺癌侵袭性强、复发率高、预后差。随着分子生物学研究的深入,肿瘤靶向治疗的研究和临床应用取得了突破性进展,HER2靶向治疗的进展改善了HER2阳性乳腺癌患者的生存。针对HER2的单克隆抗体、酪氨酸激酶抑制剂和抗体–药物偶联物的引入显著改善了HER2阳性乳腺癌患者的预后。典型的是抗HER2抗体曲妥珠单抗,已被认为是针对HER2阳性肿瘤最有效的治疗药物之一。HER2基因在乳腺癌诊断、治疗及评价预后中具有重要指导价值。本文围绕HER2阳性乳腺癌患者的抗HER2单克隆抗体的靶向治疗做一综述。

关键词

乳腺癌,人表皮生长因子受体-2,曲妥珠单抗,靶向治疗

Research Progress of Trastuzumab Targeted Therapy for HER2-Positive Breast Cancer

Shishuang Xu1,2

1The First Clinical School of Medicine, Yan’an University, Yan’an Shaanxi

2Department of Oncology, Yan’an University Affiliated Hospital, Yan’an Shaanxi

Received: Jun. 25th, 2023; accepted: Jul. 19th, 2023; published: Jul. 27th, 2023

ABSTRACT

Breast cancer is a growing trend, with human epidermal growth factor receptor 2 (HER2) positive breast cancer accounting for 15%~20% of all breast cancers. HER2 positive breast cancer is aggressive, has high recurrence rate and poor prognosis. With the deepening of molecular biology research, the research and clinical application of tumor targeted therapy have made breakthroughs, and the progress of HER2-targeted therapy has improved the survival of patients with HER2-positive breast cancer. The introduction of monoclonal antibodies against HER2, tyrosine kinase inhibitors, and antibody-drug conjugates significantly improved outcomes in patients with HER2-positive breast cancer. Typical is the anti-HER2 antibody trastuzumab, which has been recognized as one of the most effective therapies against HER2-positive tumors. HER2 gene has important guiding value in diagnosis, treatment and prognosis evaluation of breast cancer. This review focuses on the targeted therapy of anti-HER2 monoclonal antibodies in patients with HER2 positive breast cancer.

Keywords:Breast Neoplasms, HER2, Trastuzumab, Targeted Therapy

Copyright © 2023 by author(s) and Hans Publishers Inc.

This work is licensed under the Creative Commons Attribution International License (CC BY 4.0).

http://creativecommons.org/licenses/by/4.0/

1. 引言

根据2020年肿瘤数据库GLOBOCAN报告,乳腺癌是女性高发癌症,每年约有230万新发病例,平均每年导致68万人死亡。发病率和死亡率均居女性癌症的首位 [1] 。其中HER2阳性乳腺癌在乳腺癌各亚型中发病率高。在过去的二十年中,针对HER2的单克隆抗体(MoAbs)、酪氨酸激酶抑制剂(TKIs)和抗体–药物偶联物(ADCs)的引入显著改善了疾病阶段的患者预后 [2] 。2017年ESMO大会上,ExteNET研究更新了研究结果,中位随访时间5.2年,结果显示在曲妥珠单抗治疗的基础上再接受为期1年的来那替尼治疗,能够提高HER2 + 早期乳腺癌患者的5年无浸润性肿瘤复发生存率iDFS (90.2% vs 87.7%, P = 0.0083) [3] 。此外,根据MARIANNE III期试验结果显示,T-DM1单药组、T-DM1联合帕妥珠单抗组、曲妥珠单抗联合紫杉类组的中位无病生存期(PFS)分别为14.1、15.2、13.7个月,提示T-DM1有良好的抗肿瘤作用及耐受性 [4] 。其中曲妥珠单抗是一种抗HER2胞外区的人源化单克隆抗体,成为目前公认的治疗HER2阳性乳腺癌的最主要的HER2靶向治疗方法 [5] [6] 。然而,也存在一些缺点,特别是心脏毒性和耐药性,这在临床使用中引起了人们的注意。因此,了解其作用机制对建立改进的治疗策略至关重要。本文主要对曲妥珠单抗在HER2阳性乳腺癌治疗中的研究进展及应用现状作如下综述。

2. 曲妥珠单抗

2.1. 作用机理

曲妥珠单抗(Trastuzumab,商品名赫赛汀)是一种人源化单克隆抗体(免疫球蛋白G1、IgG1)。IgG1与HER2的结构域IV胞外区结合,通过上调Cdk抑制剂p27和阻断Akt和MAPK通路 [7] 引起G1期细胞阻滞,导致HER2受体的丢失从而抑制细胞的存活和生长机制。HER2与曲妥珠单抗的相互作用通过多种方式阻止酪氨酸激酶信号传导。它可以阻断HER2与其他HER受体形成二聚体,并阻断胞外区域的裂解。它还可以诱导被动内吞,从而使靶向受体被溶酶体降解 [8] 。或者,Trastuzumab的Fc区与效应免疫细胞(如自然杀伤细胞)的Fcγ受体Ⅲ结合,通过抗体依赖的细胞介导的细胞毒作用(antibody-dependent cell-mediated cytotoxicity, ADCC) [9] 杀伤肿瘤细胞。由于靶点的高度特异性,与传统化疗药物相比,细胞毒性副作用减少,因此维持了较高的生活质量 [10] 。曲妥珠单抗还可以阻止HER2异源二聚体的形成,从而下调细胞内PI3K-Akt通路。进一步通过原癌基因酪氨酸蛋白激酶Src/黏着斑激酶(Src/Fak)通路 [11] 抑制细胞存活机制。有趣的是,曲妥珠单抗已被证明具有抗血管生成特性,降低血管内皮生长因子(VEGF)表达,并可能增加血管通透性 [12] 。通过ELISA法检测治疗组和对照组肿瘤组织中VEGF-A浓度(pg/mL)。在第4天,ELISA检测显示,与对照组相比,治疗组肿瘤中的VEGF显著降低(P = 0.03) [13] 。这被提出来促进药物递送到肿瘤,但它同时与血管副作用有关。

2.2. 临床应用

针对HER2基因过表达的乳腺癌的单克隆抗体,曲妥珠单抗是全球首个获批上市治疗乳腺癌的靶向药物。曲妥珠单抗作为一种重组人单克隆抗体,其结合于HER2胞外结构域IV,抑制同二聚体的形成,从而阻断下游信号的产生 [14] 。此外,曲妥珠单抗能与人体免疫细胞作用,产生抗体依赖的细胞毒作用。曲妥珠单抗目前已广泛应用于HER2阳性乳腺癌患者的新辅助治疗、术后辅助s治疗及晚期解救治疗,均取得了良好的抗肿瘤效果。曲妥珠单抗的临床应用不仅改变了早期HER2阳性乳腺癌患者的预后,也成为HER2阳性复发转移性乳腺癌一线及疾病进展后的标准治疗方案 [15] 。在目前乳腺癌临床治疗中占据主导地位。

2.3. 局限性

但是,曲妥珠单抗有几个局限性。首先,它并不是对每一个HER2阳性的患者都有效,甚至在那些敏感的患者中,在治疗过程中,相当一部分患者往往会出现耐药。其次,它具有心脏毒性,增加了早期心力衰竭等心脏问题的风险。第三,曲妥珠单抗只靶向受体二聚体中的一个成员,从而防止信号阻断是完全的 [16] 。由于这些原因,重要的是要研究能够靶向HER2受体其他区域的药物,这些药物可以与曲妥珠单抗一起使用。

3. HER2阳性乳腺癌的研究进展

HER2阳性乳腺癌约占所有乳腺癌的15%~20% [17] [18] 。这种侵袭性疾病亚型的特征是人表皮生长因子受体2 (HER2,又称erbB2)的过度表达,典型的是通过ERBB2扩增。HER2属于受体酪氨酸激酶家族,有4个成员:HER1 (又称EGFR)、HER2、HER3和HER4。当HER蛋白被激活时,HER蛋白同源或异源二聚化,随后激活错综复杂的细胞信号级联反应,包括PI3K-AKT和RAS-MAPK (ERK)通路,调节细胞增殖和存活,以及肿瘤细胞的转移 [19] [20] 。因此,HER2是乳腺肿瘤的主要致癌驱动因子,过度表达该蛋白,从而提供了一种治疗脆弱性,可以有效地靶向HER2阳性肿瘤患者。

HER2靶向治疗的进展改善了HER2阳性乳腺癌患者的生存。局部疾病的标准治疗是化疗和1年的辅助HER2靶向治疗,典型的是抗HER2抗体曲妥珠单抗。尽管这种治疗方法有效,但仍有一部分患者会出现疾病复发 [21] 。因此,通过联合不同的HER2靶向药物或延长HER2靶向治疗的持续时间来提高治疗效果已成为人们关注的焦点。事实上,HER2双靶向治疗、延长疗程的抗HER2治疗,以及抗HER2抗体-药物偶联物T-DM1的辅助治疗,均已被批准用于临床。

目前已开发了几类抗HER2药物,包括:1) 与HER2胞外区结合的单克隆抗体,如曲妥珠单抗和帕妥珠单抗,它们通过抑制HER2信号传导发挥直接抗肿瘤作用,通过与宿主免疫系统的相互作用诱导抗体依赖的细胞毒性从而发挥间接抗肿瘤作用;2) 小分子酪氨酸激酶抑制剂(TKIs),包括拉帕替尼、奈拉替尼和阿法替尼,可与HER2及其他HER家族成员的胞内酪氨酸激酶结构域结合;3) 抗体–药物偶联物(ADCs),如曲妥珠单抗emtansine (T-DM1),由靶向HER2胞外区的单克隆抗体与细胞毒剂连接而成 [22] [23] [24] 。随着HER2阳性靶向药物的相继问世,HER2阳性乳腺癌患者的生存率和预后得到了极大的改善。但是,靶向药物具有一定耐药性。一般来说,对HER2靶向药物的耐药可归因于不同的机制,尽管有些可能是不同药物之间共有的,这些机制要么在初始治疗时流行(固有耐药),要么在治疗过程中出现,因为选择和最终主导了罕见的先前存在或新获得的耐药亚克隆(获得性耐药)。HER家族受体的不完全抑制,通过未被抑制的HER蛋白的代偿信号实现持续的信号传递,是抗HER2治疗失败的主要原因 [25] 。通过联合使用HER靶向药物,包括两种抗HER2抗体(曲妥珠单抗和帕妥珠单抗)联合使用或与强效TKI联合使用,有望克服这一不足,实现对HER家族受体的全面抑制 [26] [27] [28] 。

4. 曲妥珠单抗在HER2阳性乳腺癌的治疗进展

4.1. 术前新辅助治疗

GeparQuattro研究 [29] 证实蒽环类 + 紫杉类新辅助化疗联合曲妥珠单抗可达到较高的病理完全缓解率(PCR),且不增加早期不良反应。NOAH研究 [30] 结果也显示,与单独化疗组比较,曲妥珠单抗联合化疗组可明显提高HER2阳性患者的pCR率,并使患者死亡率和复发率分别下降了33%和40%。这一结果在炎性乳腺癌的亚组分析中也得到了验证。因此,曲妥珠单抗联合紫杉类化疗为基础的方案,成为HER2阳性乳腺癌新辅助治疗的基本方案。在曲妥珠单抗时代,15%~25%的早期HER2 + 肿瘤患者在接受曲妥珠单抗和化疗,但仍有疾病复发,这可能部分归因于HER家族受体的不完全抑制 [31] [32] 。HER2双靶向治疗的卓越疗效首次在转移性乳腺癌患者的克莱奥帕特拉试验中得到证实,在曲妥珠单抗和多西他赛的基础上加用帕妥珠单抗,获得了前所未有的15.7个月总生存(OS)获益(中位OS分别为56.5个月和40.8个月;HR 0.6)。随后,为了提高pCR率,在新辅助治疗中进行了双重HER2阻断试验,在曲妥珠单抗和化疗的基础上加用帕妥珠单抗或拉帕替尼,多个试验的结果确实显示了更高的pCR率(95% CI 0.56~0.84; P < 0.001) [33] 。KRISTINE III期临床试验显示,HER2阳性乳腺癌患者的早期新辅助治疗中,曲妥珠单抗 + 帕妥珠单抗 + 化疗方案的pCR率比T-DM1 + 帕妥珠单抗更高,但T-DM1 + 帕妥珠单抗方案的安全性更佳 [34] ,可能是与偶联于曲妥珠单抗上抑制微管形成的药物靶向于肿瘤细胞,缺乏整体抗肿瘤的效果有关,并有待于进一步研究。

4.2. 术后辅助治疗

NSABP B-31与NCCTG N9831研究联合分析显示 [32] [35] ,辅助化疗后添加曲妥珠单抗可将患者的复发风险降低一半,无病生存期(DFS)和总生存期(OS)均得到改善。曲妥珠单抗联合化疗较曲妥珠单抗序贯化疗更能提高HER2阳性早期乳腺癌患者的治疗效果。接受曲妥珠单抗辅助治疗的女性5年无病生存率为94.8%,而未接受曲妥珠单抗(P = 0.22)的女性DFS为82.7%。接受曲妥珠单抗辅助治疗的女性5年总生存率为100%,而未接受曲妥珠单抗(P = 0.038)辅助治疗的女性5年总生存率为90.4% [36] 。有临床试验证实 [37] ,术后辅助治疗中的双靶向治疗组的无病生存期与单靶向治疗组比较无明显获益,且双靶向治疗组的腹泻、皮疹等发生率反而高于单靶向治疗组,提示拉帕替尼联合曲妥珠单抗辅助治疗HER2阳性乳腺癌并不能改善生存。

4.3. 晚期治疗

Slamon等 [38] 领衔的H0648g研究首次证实了一线曲妥珠单抗联合化疗(包括蒽环类或紫杉类药物)与单独化疗相比,能提高HER2阳性晚期乳腺癌的客观缓解率(objective response, ORR) (50% vs 32%, P < 0.001),明显延长疾病进展时间(7.4个月 vs 4.6个月,P < 0.001)及OS (25.1个月 vs 20.3个月,P = 0.046)。M77001研究 [39] 进一步肯定了上述研究的结果,曲妥珠单抗联合多西他赛作为HER2阳性晚期乳腺癌患者的一线治疗优于单独使用多西他赛,并且几乎没有其他毒性。基于CLEOPATRA试验 [40] [41] ,目前推荐的HER2+晚期乳腺癌患者的一线治疗首选方案为曲妥珠单抗联合帕妥珠单抗与紫杉类。上述研究确立了曲妥珠单抗联合紫杉类在一线标准治疗的地位。此外,Robert等 [42] 还发现曲妥珠单抗、紫杉醇联合卡铂疗效优于曲妥珠单抗联合紫杉醇,能提高ORR (52% vs 36%, P = 0.04)和PFS (10.7个月 vs 7.1个月,P = 0.03)。对于HER2+的转移性乳腺癌患者,sysucc-002研究表明,曲妥珠单抗加内分泌治疗的疗效不亚于曲妥珠单抗加化疗,且毒性反应更低。

5. 总结

随着我们对HER2 + 乳腺癌的基因组特征以及分子和突变进化的了解加深,该领域不可否认地朝着精准医疗的方向发展。毫无疑问,曲妥珠单抗是HER2乳腺癌治疗的重大突破,既延长了乳腺癌患者的生命,又挽救了乳腺癌患者的生命。然而,关于最优持续时间和阻力的问题仍然需要更精确的回答。关于心脏毒性的副作用也不应该被忽视,前瞻性的有益方案应该考虑进一步减少这种副作用。展望未来,对ErbB受体下游异常的研究可以突出更多针对未来治疗剂的靶点。这些应着重于抑制增强的致瘤潜能,这可能会改善和延长寿命。尽管有这些改善,治疗抵抗的出现仍然是一个常见的和具有挑战性的事件,特别是在晚期疾病环境中,并且严重提醒我们需要更有效的治疗方法。破解耐药机制将为开发有效规避疾病复发的方法奠定基础。在HER2阳性乳腺癌患者的综合治疗中,应依据患者的具体病情,做出最有利于患者的组合治疗,最大限度的延长患者的总生存时间,做到真正意义上的个体化精准治疗。

文章引用

胥世霜. 曲妥珠单抗靶向治疗HER2阳性乳腺癌的研究进展
Research Progress of Trastuzumab Targeted Therapy for HER2-Positive Breast Cancer[J]. 临床医学进展, 2023, 13(07): 12031-12037. https://doi.org/10.12677/ACM.2023.1371686

参考文献

  1. 1. Sung, H., Ferlay, J., Siegel, R.L., et al. (2021) Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA, 71, 209-249. https://doi.org/10.3322/caac.21660

  2. 2. Vernieri, C., Milano, M., Brambilla, M., Mennitto, A., Maggi, C., Cona, M.S., Prisciandaro, M., Fabbroni, C., Celio, L., Mariani, G., Bianchi, G.V., Capri, G. and de Braud, F. (2019) Resistance Mechanisms to Anti-HER2 Therapies in HER2-Positive Breast Cancer: Current Knowledge, New Research Directions and Therapeutic Perspectives. Critical Reviews in Oncology/Hematology, 139, 53-66. https://doi.org/10.1016/j.critrevonc.2019.05.001

  3. 3. Martin, M., Holmes, F.A., Ejlertsen, B., et al. (2017) Nerat-inib after Trastuzumab-Based Adjuvant Therapy in HER-2 Positive Breast Cancer (ExteNET): 5-Year Analysis of a Randomised, Double Blind, Placebo-Controlled, Phase 3 Trim. The Lancet Oncology, 18, 1688-1700. https://doi.org/10.1016/S1470-2045(17)30717-9

  4. 4. Ellis, P.A., Barrios, C.H., Eiermann, W., et al. (2015) Phase III, Randomized Study of Trastuzumab Emtansine (T -DMI) ± Pertuzumab (P) vs Trastuzumab + Taxane(HT) for First-Line Treatment of HER2-Positive MBC: Primary Results from the MARIANNE Study. Journal of Clinical On-cology, 33, 507. https://doi.org/10.1200/jco.2015.33.15_suppl.507

  5. 5. Cardoso, F., Kyriakides, S., Ohno, S., et al. (2019) Early Breast Cancer: ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Journal of Clinical Oncol-ogy, 30, 1194-220. https://doi.org/10.1093/annonc/mdz173

  6. 6. Cardoso, F., Senkus, E., Costa, A., et al. (2018) 4th ESO-ESMO International Consensus Guidelines for Advanced Breast Cancer (ABC 4). Annals of Oncology, 29, 1634-1657. https://doi.org/10.1093/annonc/mdy192

  7. 7. Le, X.F., Pruefer, F. and Bast Jr., R.C. (2005) HER2-Targeting Antibodies Modulate the Cyclin-Dependent Kinase Inhibitor p27Kip1 via Multiple Signaling Pathways. Cell Cycle, 4, 87-95. https://doi.org/10.4161/cc.4.1.1360

  8. 8. Austin, C.D., De Maziere, A.M., Pisacane, P.I., van Dijk, S.M., Eigenbrot, C., Sliwkowski, M.X., Klumperman, J. and Scheller, R.H. (2004) Endocytosis and Sorting of ErbB2 and the Site of Action of Cancer Therapeutics Trastuzumab and Geldanamycin. Molecular Biology of the Cell, 15, 5268-5282. https://doi.org/10.1091/mbc.e04-07-0591

  9. 9. Collins, D.M., O’Donovan, N., McGowan, P.M., O’Sullivan, F., Duffy, M.J. and Crown, J. (2012) Trastuzumab Induces Antibody-Dependent Cell-Mediated Cytotoxicity (ADCC) in HER-2-Non-Amplified Breast Cancer Cell Lines. Annals of Oncology, 23, 1788-1795. https://doi.org/10.1093/annonc/mdr484

  10. 10. Osoba, D., Slamon, D.J., Burchmore, M. and Murphy, M. (2002) Ef-fects on Quality of Life of Combined Trastuzumab and Chemotherapy in Women with Metastatic Breast Cancer. Journal of Clinical Oncology, 20, 3106-3113. https://doi.org/10.1200/JCO.2002.03.090

  11. 11. Xu, Y., Benlimame, N., Su, J., He, Q. and Alaoui-Jamali, M.A. (2009) Regulation of Focal Adhesion Turnover by ErbB Signalling in Invasive Breast Cancer Cells. British Journal of Cancer, 100, 633-643. https://doi.org/10.1038/sj.bjc.6604901

  12. 12. Petit, A.M., Rak, J., Hung, M.C., Rockwell, P., Goldstein, N., Fendly, B. and Kerbel, R.S. (1997) Neutralizing Antibodies against Epidermal Growth Factor and ErbB-2/neu Receptor Tyrosine Kinases Downregulate Vascular Endothelial Growth Factor Production by Tumor Cells in Vitro and in Vivo: Angiogenic Implications for Signal Transduction Therapy of Solid Tumors. Am. J. Pathol, 151, 1523-1530.

  13. 13. Sorace, A.G., Quarles, C.C., Whisenant, J.G., Hanker, A.B., McIntyre, J.O., Sanchez, V.M. and Yankeelov, T.E. (2016) Trastuzumab Improves Tumor Perfusion and Vascular Delivery of Cytotoxic Therapy in a Murine Model of HER2+ Breast Cancer: Preliminary Results. Breast Cancer Research and Treatment, 155, 273-284. https://doi.org/10.1007/s10549-016-3680-8

  14. 14. 王慧, 赵安帝, 杨谨. 曲妥珠单抗联合帕妥珠单抗用于HER2阳性早期乳腺癌研究进展[J]. 华中科技大学学报(医学版), 2020, 49(1): 111-116.

  15. 15. 韩萌萌, 冯雪园, 马宁. 人表皮生长因子受体2阳性乳腺癌的靶向治疗研究进展[J]. 中华普通外科学文献(电子版), 2021, 15(6): 453-458.

  16. 16. Wang, Q., Zhang, X., Shen, E., Gao, J., Cao, F., Wang, X., Li, Y., Tian, T., Wang, J., Chen, Z., Wang, J. and Shen, L. (2016) The Anti-HER3 Antibody in Combination with Trastuzumab Exerts Synergistic Antitumor Activity in HER2- Positive Gastric Cancer. Cancer Letters, 380, 20-30. https://doi.org/10.1016/j.canlet.2016.06.005

  17. 17. Slamon, D.J., et al. (1989) Studies of the HER-2/neu Protoonco-gene in Human Breast and Ovarian Cancer. Science, 244, 707-712. https://doi.org/10.1126/science.2470152

  18. 18. Wolff, A.C., et al. (2013) Recommendations for Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Update. Journal of Clinical Oncology, 31, 3997-4013. https://doi.org/10.1200/JCO.2013.50.9984

  19. 19. Rimawi, M.F., Schiff, R. and Osborne, C.K. (2015) Targeting HER2 for the Treatment of Breast Cancer. Annual Review of Medicine, 66, 111-128. https://doi.org/10.1146/annurev-med-042513-015127

  20. 20. Moasser, M.M. (2007) The Oncogene HER2: Its Sig-naling and Transforming Functions and Its Role in Human Cancer Pathogenesis. Oncogene, 26, 6469-6487. https://doi.org/10.1038/sj.onc.1210477

  21. 21. Goutsouliak, K., Veeraraghavan, J., Sethunath, V., De Angelis, C., Osborne, C.K., Rimawi, M.F. and Schiff, R. (2020) Towards Personalized Treatment for Early Stage HER2-Positive Breast Cancer. Nature Reviews Clinical Oncology, 17, 233-250. https://doi.org/10.1038/s41571-019-0299-9

  22. 22. Clynes, R.A., Towers, T.L., Presta, L.G. and Ravetch, J.V. (2000) Inhibitory Fc Receptors Modulate in Vivo Cytotoxicity against Tumor Targets. Nature Medicine, 6, 443-446. https://doi.org/10.1038/74704

  23. 23. Goutsouliak, K., Veeraraghavan, J., Sethunath, V., De Angelis, C., Osborne, C.K., Rimawi, M.F. and Schiff, R. (2020) Towards Personalized Treatment for Early Stage HER2-Positive Breast Cancer. Nature Reviews Clinical Oncology, 17, 233-250. https://doi.org/10.1038/s41571-019-0299-9

  24. 24. Scheuer, W., et al. (2009) Strongly Enhanced Antitumor Activity of Trastuzumab and Pertuzumab Combination Treatment on HER2-Positive Human Xenograft Tumor Models. Cancer Research, 69, 9330-9336. https://doi.org/10.1158/0008-5472.CAN-08-4597

  25. 25. Yamashita-Kashima, Y., et al. (2011) Pertuzumab in Com-bination with Trastuzumab Shows Significantly Enhanced Antitumor Activity in HER2-Positive Human Gastric Cancer Xenograft Models. Clinical Cancer Research, 17, 5060- 5070. https://doi.org/10.1158/1078-0432.CCR-10-2927

  26. 26. Gianni, L., et al. (2012) Efficacy and Safety of Neoadjuvant Pertuzumab and Trastuzumab in Women with Locally Advanced, Inflammatory, or Early HER2-Positive Breast Cancer (NeoSphere): A Randomised Multicentre, Open-Label, Phase 2 Trial. The Lancet Oncology, 13, 25-32. https://doi.org/10.1016/S1470-2045(11)70336-9

  27. 27. Llombart-Cussac, A., et al. (2017) HER2-Enriched Subtype as a Predictor of Pathological Complete Response Following Trastuzumab and Lapatinib without Chemotherapy in Ear-ly-Stage HER2-Positive Breast Cancer (PAMELA): an Open-Label, Single-Group, Multicentre, Phase 2 Trial. The Lan-cet Oncology, 18, 545–554. https://doi.org/10.1016/S1470-2045(17)30021-9

  28. 28. Rimawi, M.F., et al. (2013) Multicenter Phase II Study of Neoadjuvant Lapatinib and Trastuzumab with Hormonal Therapy and without Chemotherapy in Patients with Human Epidermal Growth Factor Receptor 2-Overexpressing Breast Cancer: TBCRC 006. Journal of Clinical Oncology, 31, 1726-1731. https://doi.org/10.1200/JCO.2012.44.8027

  29. 29. Untch, M., Rezai, M., Loibl, S., et al. (2010) Neoad-juvant Treatment with Trastuzumab in HER2-Positive Breast Cancer: Results from the GeparQuattro Study. Journal of Clinical Oncology, 28, 2024-2031. https://doi.org/10.1200/JCO.2009.23.8451

  30. 30. Gianni, L., Eiermann, W., Semiglazov, V., et al. (2010) Neoadju-vant Chemotherapy with Trastuzumab Followed by Adjuvant Trastuzumab versus Neoadjuvant Chemotherapy Alone, in Patients with her2positive Locally Advanced Breast Cancer (the NOAH trial): A Randomised Controlled Superiority Tri-al with a Parallel HER2negative Cohort. The Lancet, 375, 377-384. https://doi.org/10.1016/S0140-6736(09)61964-4

  31. 31. Cameron, D., et al. (2017) 11 Years’ Follow-Up of Trastuzumab after Adjuvant Chemotherapy in HER2-Positive Early Breast Cancer: Final Analysis of the HERceptin Ad-juvant (HERA) Trial. The Lancet, 389, 1195-1205. https://doi.org/10.1016/S0140-6736(16)32616-2

  32. 32. Perez, E.A., et al. (2014) Trastuzumab Plus Adjuvant Chem-otherapy for Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer: Planned Joint Analysis of Overall Survival from NSABP B-31 and NCCTG N9831. Journal of Clinical Oncology, 32, 3744-3752. https://doi.org/10.1200/JCO.2014.55.5730

  33. 33. Swain, S.M., et al. (2015) Pertuzumab, Trastuzumab and Docet-axel in HER2-Positive Metastatic Breast Cancer. The New England Journal of Medicine, 372, 724-734. https://doi.org/10.1056/NEJMoa1413513

  34. 34. Hurvitz, S.A., Martin, M., Symmans, W.F., et al. (2018) Neoadju-vant Trastuzumab, Pertuzumab and Chemotherapy versus Trastuzumab Emtansine Plus Pertuzumab in Patients with HER-2-Positive Breast Cancer (KRISTINE): A Randomised, Open-Label, Multicentre, Phase 3 Trial. The Lancet On-cology, 19, 115-126. https://doi.org/10.1016/S1470-2045(17)30716-7

  35. 35. Gallagher, C.M., More, K., Masaquel, A., et al. (2016) Sur-vival in Patients with Non-Metastatic Breast Cancer Treated with Adjuvant Trastuzumab in Clinical Practice. Springer-plus, 5, Article No. 395. https://doi.org/10.1186/s40064-016-2008-9

  36. 36. Ali, S., Hendry, J., Le, D., Mondal, P.K., Sami, A., Chalchal, H., Haider, K., Ahmed, O., El-Gayed, A., Wright, P., Pauls, M., Johnson, K. and Ahmed, S. (2022) Efficacy of Adjuvant Trastuzumab in Women with HER2-Positive T1a or bN0M0 Breast Cancer: A Population-Based Cohort Study. Scientific Reports, 12, Article No. 1068. https://doi.org/10.1038/s41598-022-05209-8

  37. 37. Piccart-Gebhart, M., Holmes, E., Baselga, J., et al. (2016) Adju-vant Lapatinib and Trastuzumab for Early Human Epidermal Growth Factor Receptor 2positive Breast Cancer: Results from the Randomized Phase III Adjuvant Lapatinib and/or Trastuzumab Treatment Optimization Trial. Journal of Clinical Oncology, 34, 1034-1042. https://doi.org/10.1200/JCO.2015.62.1797

  38. 38. Slamon, D.J., Leyland-jones, B., Shak, S., et al. (2001) Use of Chemotherapy plus a Monoclonal Antibody against HER2 for Metastatic Breast Cancer that Overexpresses HER2. The New England Journal of Medicine, 344, 783-792. https://doi.org/10.1056/NEJM200103153441101

  39. 39. Marty, M., Cognetti, F., Maraninchi, D., et al. (2005) Ran-domized Phase Iitrial of the Efficacy and Safety of Trastuzumab Combined with Docetaxel in Patients with Human Epi-dermal Growth Factor Receptor 2-Positive Metastatic Breast Cancer Administered as First-Line Treatment: The M77001 Study Group. Journal of Clinical Oncology, 23, 4265-4274. https://doi.org/10.1200/JCO.2005.04.173

  40. 40. Giordano, S.H., Temin, S., Kirshner, J.J., et al. (2014) Systemic Therapy for Patients with Advanced Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer: American So-ciety of Clinical Oncology Clinical Practice Guideline. Journal of Clinical Oncology, 32, 2078-2099. https://doi.org/10.1200/JCO.2013.54.0948

  41. 41. Bachelot, T., Ciruelos, E., Schneeweiss, A., et al. (2019) Prelimi-nary Safety and Efficacy of First-Line Pertuzumab Combined with Trastuzumab and Taxane Therapy for HER2-Positive Locally Recurrent or Metastatic Breast Cancer (PERUSE). Annals of Oncology, 30, 766-773. https://doi.org/10.1093/annonc/mdz061

  42. 42. Robert, N., Leyland-jones, B., Asmar, L., et al. (2006) Randomized Phase III Study of Trastuzumab, Paclitaxel and Carboplatin Compared with Trastuzumab and Paclitaxel in Women with HER-2-Overexpressing Metastatic Breast Cancer. Annals of Oncology, 24, 2786-2792. https://doi.org/10.1200/JCO.2005.04.1764

期刊菜单