HER-2低表达乳腺癌的研究进展与未来展望
Research Progress and Future Prospect of HER-2 Low Expression Breast Cancer
摘要: 近年来对HER-2低表达乳腺的关注,在全球范围日益增加,随着新型靶向药物的研发及检测技术的发展,HER-2低表达乳腺癌与其他类型乳腺癌表现出不同的病理学及临床特征。对于HER-2低表达乳腺癌患者的预后,众多学者持有不同的观点尚无统一论据。本文阐述了近年来HER-2低表达乳腺癌的最新研究进展,重点介绍了HER-2低表达乳腺癌的生物学特性、目前病理实践中免疫组化判定HER-2低表达乳腺癌所遇到的问题,以及对这一新型领域未来的发展做一概述。
Abstract: In recent years, HER-2 low expression breast cancer has received increasing attention. With the development of new HER-2 targeted drugs and detection methods, HER-2 low breast cancer exhibit pathological and clinical features that differ from other types of breast cancer. For the prognosis of HER-2-low expression breast cancer, many scholars hold different views, and there is no unified ev-idence. In this paper, the latest research progress in breast cancer with low HER-2 expression in recent years is reviewed, focusing on the biological characteristics of HER-2 low breast cancer, and the problems encountered in the immunohistochemical detection of HER-2 low expression breast cancer in current pathological practice, and the future development of this new field is summarized.
文章引用:买吐鲁佰·米尔扎合买提, 卡吾力·居买, 地力木拉提·艾斯木吐拉, 李慧芳, 阿力清·阿不都萨拉木. HER-2低表达乳腺癌的研究进展与未来展望[J]. 临床医学进展, 2023, 13(10): 16593-16601. https://doi.org/10.12677/ACM.2023.13102323

1. 引言

乳腺癌是女性发病率在全球范围居于首位的恶性肿瘤,且在女性常见恶性肿瘤中死亡率为第二位 [1] 。人表皮生长因子2 (human epidermal growth factor receptor 2, HER-2)从评定乳腺癌预后的生物标志物到肿瘤患者靶向药物的应用发挥着重要作用。HER-2是一种跨膜酪氨酸激酶受体蛋白,由位于17号染色体长臂上的ERBB2基因编码。正常乳腺上皮细胞在每条17号染色体上有一个HER-2基因拷贝,每个细胞表达约20,000个HER-2受体 [2] 。当HER-2基因扩增时,肿瘤细胞表面的HER-2受体分子显著增加,约为200万个,扩增的HER-2会在二聚化时刺激其内在激酶活性,导致细胞内信号级联的激活,最终引起细胞增殖、血管生成、侵袭和转移。根据癌组织中HER-2表达水平、激素受体水平(hormone receptor, HR)及Ki-67表达水平乳腺癌分为以下四种不同的分子分型:HER-2阳性型、三阴型(triple negtive breast cancer, TNBC)、Luminal A型和Luminal B型。临床上,随着HER-2靶向药物的发展,从单克隆抗体曲妥珠单抗开始,随后是帕妥珠单抗、酪氨酸激酶抑制剂(如:拉帕替尼、纳拉替尼和图卡替尼),以及最近的抗体药物偶联物ADC (antibody-drug conjugate, ADC),使HER-2阳性乳腺癌患者从中获益 [3] 。随着检测技术与药物的开发,研究者就HER-2低表达乳腺癌患者能否从抗HER-2治疗上获益而展开了相关试验 [4] [5] 。下面将以近年来HER-2低表达乳腺癌的诊断、临床病理特征、治疗以及这一新兴乳腺癌的未来发展方向作一综述。

2. HER-2低表达乳腺癌的诊断方式

正确检测人类表皮生长因子受体2 (HER-2)在乳腺肿瘤组织中的表达水平和基因扩增状态,对判断乳腺癌患者预后及评定临床疗效有重要影响。目前检测乳腺恶性肿瘤中HER-2表达水平的方法有两种:检测HER-2蛋白表达水平用免疫组化法(immunohistochemistry, IHC),检测HER-2基因扩增水平应用原位杂交法(in situ hybridization, ISH)。ISH包括荧光原位杂交(fluorescence in situ hybridization, FISH)和亮视野原位杂交。常用的亮视野原位杂交方法有显色原位杂交(chromogenic in situ hybridization, CISH) [6] [7] 和银增强原位杂交(silver-enhanced in situ hybridization, SISH) [8] [9] 。2018年版美国临床肿瘤学会(American Society of Clinical Oncology, ASCO)和美国病理学家学会(College of American Pathologists, CAP) HER-2检测指南(ASCO/CAP指南) [10] ,将IHC结果为HER-2强阳性(3+)或中阳性(2+)伴有FISH结果提示基因过度扩增(+),认为是HER-2阳性乳腺癌。IHC检测结果为(0)、(1+)或(2+)并伴FISH基因无扩增,认为HER-2阴性乳腺癌。其中,约10%~20%的肿瘤为HER-2阳性,80%~90%为HER-2阴性 [11] 。随着检测技术的进步以及抗HER-2靶向药物的应用,使全球众多学者的注意力转移到HER-2低表达这新亚型乳腺癌上。2021年版《乳腺癌诊疗指南》第一次细化HER-2阴性乳腺癌临床亚型。把IHC结果为(2+)且FISH结果显示基因未扩增,或IHC结果为(1+)的乳腺癌划分为HER-2低表达乳腺癌。这一新型乳腺癌占所有乳腺癌患者的45%~55% [12] 。传统的根据HER-2表达水平的二分类概念(即阳性与阴性)到现在提出的三分类概念,给HER-2表达水平的检测及免疫组化结果鉴定的准确性及可靠性带来新的挑战。其中提高病理医师判读检测结果的一致性成为主要的问题之一 [13] 。根据我国2019版《乳腺癌HER-2检测指南》 [14] ,无着色或≤10%的浸润癌细胞呈现不完整的、微弱的细胞膜染色为IHC(0),>10%的浸润癌细胞呈现不完整的、微弱的细胞膜染色为IHC(1+)。在病理学家判读过程中,对比IHC(0)与IHC(1+)两组,一些研究发现有不一致的情况 [15] [16] [33] 。

Schettini等人对5名乳腺癌专科病理学家判读100例样本进行了HER-2染色的病理间一致性分析。该研究结果显示,100例患者中有35例IHC评分不一致 [17] 。另外一项研究提示,18名病理学家在不知道研究目的的情况下,阅读了一组选定的乳腺癌组织切片,最后结果中,HER-2 (0)和HER-2 (1+)之间只有26.0%的一致性,而HER (2+)和HER-2 (3+)之间的一致性为58.0% [16] 。以上研究结果表明,判读免疫组化过程中,病理学家之间判读染色结果的一致性面临着挑战。

尽管有标准化的HER-2免疫组化检测指南,但结果可能受到其他因素的影响,这其中包括肿瘤细胞HER-2表达随时间的变化。韩国一项回顾性研究结果显示,新辅助化疗前后免疫组化结果有统计学差异。10.3%的HR阳性并且HER2阴性的乳腺癌倾向于转化为三阴性(TNBC)乳腺癌,而三阴性(TNBC)乳腺癌转化为HR阳性且HER2阴性的肿瘤的百分比为34.6% [18] 。分子亚型改变,表明肿瘤预后也将发生改变。其他影响因素还包括肿瘤异质性或个体实验室和读者之间的不一致等。

3. HER-2低表达乳腺癌的临床及病理特征

临床上,通常HER-2低表达乳腺癌包含在Luminal型或三阴性(TNBC)乳腺癌当中。与HER-2阴性乳腺癌相比,HER-2低表达乳腺癌是否代表了乳腺癌中一个独特的生物学亚型,目前仍存在争议 [17] [20] [21] [22] [23] 。当然也有学者发现HER-2低表达乳腺癌表现出不一样的病理及临床特征 [17] [19] 。HER-2低表达乳腺癌中,高达65%~83%为HR阳性乳腺癌,其余为HR阴性肿瘤,并且HR阳性的HER-2低表达患者以Luminal型为主,HR阳性的HER-2阴性乳腺癌则以TNBC为主 [25] [26] 。因此在学者们看来,研究HER-2低表达乳腺癌的生物学行为时,HR表达状态是重要影响因素 [20] 。研究显示,HR 阳性的HER-2低表达和HER-2阴性乳腺癌患者的生存结局相似 [17] [26] [27] 。但也有学者荟萃分析结果提示,在早期乳腺癌中,无论HR状态如何,与HER-2 阴性乳腺癌相比,HER-2低表达乳腺有着更好的DFS和OS [28] 。但HER-2低表达乳腺癌原发肿瘤大小更大,淋巴结受累更多并且发病年龄较高;Ki67增值指数与组织病理学分级往往较高 [15] [25] [29] ;中枢神经系统和内脏并发症少 [30] 。除此之外,几项研究表明,与HER-2(0)乳腺癌相比,HER-2低表达乳腺癌在超重(即体重指数大于或等于25 kg/m2)的患者中更常见,且腋窝淋巴结转移率增加,有较高的IV肿瘤发病率和更高的组织学分级 [17] [24] [57] 。Pinhel等人发现HER-2表达水平则与雌激素受体(estrange receptor)表达水平呈正相关 [31] 。基因表达方面,在HR阳性的HER-2低表达乳腺癌病例中,ERBB2表达最高。增殖相关基因(如CCNE1、MKI67和EXO1)表达相对较低,管腔相关基因(ESR1、AR和BCL2)表达较高 [15] 。HER低表达乳腺癌的PIK3CA (磷脂酰肌醇3-激酶催化亚单位phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha, (PIK3CA))信号通路相关基因往往比HER-2阴性肿瘤(24.8% vs 16.3%)的突变率高;TP53突变发生率低(33.4%对44.0%) [17] [32] 。Agostinetto等人在对804例患者的回顾性分析中发现,即使与HR状态配对,HER-2低表达乳腺癌患者之间的DFS和OS也没有显著差异 [26] 。Horisawa等人也报道,在4918例患者中,无论HR状态如何,HER-2低表达和HER-2 阴性患者的预后无统计学差异 [33] 。另有研究显示,因靶向药物的应用,HER-2阳性乳腺癌患者DFS优于HER-2低表达乳腺癌患者。并且在低表达患者中,HER-2 ICH(2+)/FISH无扩增的患者的DFS较高 [29] 。基于这些相互矛盾的结果,目前还不能得出关于HER-2低表达乳腺癌预后的明确结论,这可能是由于患者群体、研究设计或随访时间的差异。如果没有包括HR状态和治疗方案的前瞻性研究,关于HER-2低表达乳腺癌患者预后的结果,可能将难以定论。

4. HER-2低表达乳腺癌的治疗进展与预后

以往的抗HER-2治疗主要针对HER-2过表达乳腺癌。但在HER-2低表达乳腺癌细胞膜上存在50万~100万个HER-2受体分子 [17] ,因此学者们考虑到抗HER-2治疗能否让HER-2低表达乳腺癌患者从中获益。自1997年FDA批准曲妥珠单抗是第一个抗HER-2靶向治疗的药物以来,我们踏入了分子靶向治疗的时代 [34] 。曲妥珠单抗与帕托珠单抗是HER-2单克隆抗体,其中前者曲妥珠单抗在体内和体外对乳腺肿瘤均有活性,且与化疗联合使用具有协同作用 [34] ,是HER-2阳性转移性乳腺癌(MBC)的一线药物。曲妥珠单抗除了阻断HER-2生长信号通路外,还会引起抗体依赖性细胞毒性(ADCC),在微转移瘤发挥更大的作用。III期NSA-B47试验显示,早期HER-2低表达乳腺癌行单独化疗与联合应用曲妥珠单抗,3270例患者侵袭性无病生存期、远处无复发生存期无明显差异。该试验结果表明低水平的HER-2表达可能不足以诱导曲妥珠单抗产生免疫反应 [27] 。

帕妥珠单抗与曲妥珠单抗相比,即使在没有HER-2过表达的情况下,也能够抑制异种移植模型的肿瘤生长 [35] 。尽管如此,在一项II期临床试验中,对HER-2阴性或低表达的乳腺肿瘤患者单独给予帕妥珠单抗后的疗效令研究者有所失望 [36] 。

ADC (antibody-drug conjugate, ADC)是一种高度特异性抗癌药物。通过将单克隆抗体与细胞毒性药物结合(称为药物有效载荷)成混合分子。它具备了抗体结合特定靶点的优势和化疗药物的细胞毒性 [37] ,从而提高选择性和疗效。因这种特殊药物机理,不仅治疗HER-2阳性癌症患者,更是让不能从常规抗HER-2治疗收益的HER-2低表达乳腺癌患者获得了更好的疗效 [38] 。曲妥珠单抗-德鲁西替康和曲妥珠单抗-杜卡玛嗪的非随机试验结果表明,在晚期HER2低表达的乳腺癌患者中有一定的疗效,客观缓解率在32%至37%之间 [4] [5] 。目前,第二代ADC药物恩美曲妥珠单抗T-DM1 (ado-trastuzumab emtansine)已用于临床治疗乳腺癌,它是由曲妥珠单抗和美坦新组成。T-DM1与肿瘤细胞表面HER-2受体结合后,一部分受体通过受体内吞过程内化,导致细胞内释放活性形式的DM1,导致肿瘤细胞死亡 [39] 。III期试验EMILIA研究结果显示,与卡培他滨 + 拉帕替尼相比,T-DM1显著改善了患者的总生存期(OS)和无进展生存期(PS),客观反应率(ORR)和中位缓解持续时间(DoR)结果也是让人欣慰,巩固了T-DM1在晚期HER-2阳性乳腺癌的二线治疗地位 [40] 。但对于低表达乳腺癌,缺少有关TDM-1的前瞻性研究。

T-DXd (Trastuzumab Deruxtecan DS-8201a) 是一种与拓扑异构酶I抑制剂偶联的HER-2靶向单克隆抗体,与SYD985代表了第三代ADC药物。T-DXd具有高效、新颖的有效载荷以及更高的抗体药物比(DAR为8:1)。肿瘤选择性可切割连接体,以及较短的有效载荷半衰期。确保更高浓度细胞毒性药物在肿瘤部位释放,避免全身暴露。DXd的高膜透性使其能够进入肿瘤微环境(tumor microenvironment, TME)中的抗原阴性肿瘤细胞 [41] 。这种直接杀伤靶向抗原阳性细胞,同时杀伤邻近的抗原阴性肿瘤细胞的现象被称为旁观者杀伤效应 [42] 。因此在杀死HER-2过表达的肿瘤细胞时,也可以杀伤HER-2阴性或低表达的肿瘤细胞。针对T-DXd在HER-2低表达乳腺癌患者的人体Ib期试验显示,54名晚期HER-2低表达乳腺癌患者应用推荐剂量的T-Dxd,总缓解率(ORR)为37%,中位无进展生存期(mPFS)为11.1个月,中位总生存期(mOS)为29.4个月。值得注意的是其中3名患者出现了T-DXd诱导的间质性肺疾病(ILD)。但T-DM1、曲妥珠单抗和伊立替康的ILD发生率分别为1.2%、0.2%~0.5%和0.9% [4] [43] 。因此需要进一步监测IDL发生概率,早期发现并确定T-Dxd的安全性及对患者的预后的影响。SYD985的有效载荷是duocarmycin,通过可切割连接子(vc-seco-DUBA)与曲妥珠单抗结合。DRA为2.8:1,尽管DRA较低,SYD985与T-DM1相比,在HER-2低乳腺癌患者的异种移植乳腺癌模型的研究中,明显比T-DM1更有效 [44] 。SYD985其首次人体试验,局部晚期肿瘤或晚期肿瘤患者中,有47例HER-2低表达乳腺癌患者。32例HR阳性患者中有9例(28%, 95% CI 13.8~46.8),15例HR阴性患者中有6例(40%, 95% CI 16.3~67.6),获得部分缓解 [5] 。

我国研发的ADC药物——维迪西妥单抗(RC48),在具备旁观者效应的同时,还具备了安全性好,不良反应更少等优点 [45] 。2021年6月,迪西他单维多汀在中国获得首个生物制剂许可申请(BLA)批准,用于治疗接受过至少两种全身化疗方案的晚期或转移性胃癌(包括胃食管交界处腺癌)。RC48抗体部分为hertuzumab,较曲妥珠单抗对HER-2靶点有更好的亲和力。连接体则是是稳定的缬氨酸–瓜氨酸(VC)连接体,只有当RC48被内吞入溶酶体时,才能被组织蛋白酶切割,从而释放有效载荷,即细胞毒性药物甲基澳瑞他汀E(MMAE)杀死目标癌细胞 [46] 。该药物对HER-2的结合特异性不受与MMAE结合的影响。在ASCO公布的70例HER-2 + BC患者和48例HER-2低表达乳腺癌患者的试验结果显示,31.4%的HER-2阳性乳腺癌患者达到了31.4%的ORR,而中位PFS为5.8个月。RC48在HER-2阳性和低表达阳性患者中均能取得良好的疗效 [47] 。RC48可能是一种很有前景的药物,关于RC48治疗转移性乳腺癌的Ib期和II/III期临床试验正在中国开展中。

XMT-1522使用HT-19代替曲妥珠单抗,HT-19结合与曲妥珠单抗不同的HER-2表位,通过可生物降解的半胱氨酸连接到另一种抗微管蛋白药物auristatin衍生物。XMT-1522在ADC药物中具有最高的DAR (12:1),引发了可控的旁观者杀伤效应,并且在HER-2阳性和低表达异种移植的癌症模型中表现优于T-DM1 [48] 。尽管有初步的临床疗效迹象,但由于商业原因,XMT-1522的开发在I期停止 [49] 。除上述的抗HER-2药物,还有ARX788、PF-06804103、MM-302、MEDI4276、A166、BAT8001、DHES0815A、ALT-P7、HER-2疫苗(Nelipepimut-S, NP-S)等。部分药物还在进行相应临床试验和探索。ADC药物的完善,将会对乳腺癌患者精准化治疗具有深刻意义。在临床试验数据集中,进一步研究患者对新型ADC的反应与HER-2表达之间的关系,也将非常有助于更好地定义HER-2低水平乳腺癌。在我们看来,相信HER-2低表达乳腺癌的定义将根据新药的开发、未来临床试验的结果、更敏感和可靠的检测方法的发展以及我们对HER-2低表达乳腺癌的理解而继续发展。

5. 展望

上述的众多研究结果与治疗进展表明,HER-2低表达乳腺癌将是一个独特类别,具有新的靶向治疗意义 [50] [51] 。标准的免疫组化对HER-2低表达的定义至关重要。测定HER-2表达水平的定量方法必定是第一步更是关键一步。未来的HER-2检测很可能将重点放到区分真阴性与HER-2低表达的研究中,甚至是超低表达(HER-2 IHC > 0, <1+)等。由于免疫组化法与荧光原位杂交法在识别低水平HER-2表达方面的潜在局限性,提高准确检测低水平HER-2表达能力,可使患者受益于新的抗HER-2药物。开发可靠和准确的定量实验室方法来确定HER-2低乳腺癌也在积极研究中。美国学者联用免疫亲和富集与多反应监测–质谱(immunoaffinity-enrichment coupled to multiple reaction monitoring-mass spectrometry immuno-MRM- MS)的方法,定量测量了冷冻和福尔马林固定石蜡包埋(FFPE)的乳腺癌活检组织中的HER-2表达水平。结果表明,该测量方法具有可接受性,即使在低表达水平下也能精确、相对定量地测定HER-2 [52] 。实时荧光定量聚合酶链反应(real-time fluorescence quantitative polymerase chain reaction, RTFQ-PCR)也是检测HER-2的方法之一 [53] 。Xu等人发现免疫组化方法测量HER-2表达的动态范围是相对有限的,特别是在HER-2阴性患者中,而基于RNA丰度的表达范围表明,定义HER-2低表达乳腺癌的分子方法可能更好地满足这一群体的治疗需求 [54] 。但RNA丰度识别HER-2低表达乳腺癌和预测治疗反应的有效性需要通过前瞻性临床试验进一步评估。先前报道的其他测定方法还包括链亲和素包被的荧光粉集成点荧光纳米颗粒(pid) [55] 、基于免疫荧光的AQUA(蛋白质表达的定量方法(enzyme-linked immunosorbent assay or AQUA酶联免疫吸附试验或AQUA)自动定量分析方法 [56] 。然而,在批准用于临床之前,任何新的HER-2蛋白检测定量方法还需要经过广泛的分析和临床验证,以证明临床效用。

尽管众多学者在HER-2低表达乳腺癌的研究上做出了巨大努力,但关于其预后及临床病理特征仍未被阐明。此外,到目前为止,没有确凿的证据支持HER-2低表达在乳腺癌中作为一个独立的预后因素和一个独特的亚型。通过更多临床试验数据,进一步深入研究患者对新型ADC药物的反应与HER-2表达水平之间的关系,也将非常有助于更好地定义HER-2低表达乳腺癌。并能帮助更多肿瘤患者获益。

NOTES

*通讯作者。

参考文献

[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: A Cancer Journal for Clinicians, 71, 209-249.
https://doi.org/10.3322/caac.21660
[2] Schechter, A.L., Hung, M.C., et al. (1985) The Neu Gene: An erbB-Homologous Gene Distinct from and Unlinked to the Gene Encoding the EGF Receptor. Science, 229, 976-978.
https://doi.org/10.1126/science.2992090
[3] Xu, Y., Wang, Y., Gong, J., et al. (2021) Phase I Study of the Re-combinant Humanized Anti-HER2 Monoclonal Antibody-MMAE Conjugate RC48-ADC in Patients with HER2-Positive Advanced Solid Tumors. Gastric Cancer, 24, 913-925.
https://doi.org/10.1007/s10120-021-01168-7
[4] Modi, S., Park, H., Murthy, R.K., et al. (2020) Antitumor Activ-ity and Safety of Trastuzumab Deruxtecan in Patients with HER-2-Low-Expressing Advanced Breast Cancer: Results from a Phase Ib Study. Journal of Clinical Oncology, 38, 1887-1896.
https://doi.org/10.1200/JCO.19.02318
[5] Banerji, U., van Herpen, C.M.L., Saura, C., et al. (2019) Trastuzumab Duocarmazine in Locally Advanced and Metastatic Solid Tumours and HER-2-Expressing Breast Cancer: A Phase 1 Dose-Escalation and Dose-Expansion Study. The Lancet Oncology, 20, 1124-1135.
https://doi.org/10.1016/S1470-2045(19)30328-6
[6] Hanna, W.M. and Kwok, K. (2006) Chromogenic In-Situ Hybridization: A Viable Alternative to Fluorescence In-Situ Hybridization in the HER-2 Testing Algorithm. Modern Pa-thology, 19, 481-487.
https://doi.org/10.1038/modpathol.3800555
[7] 张瑰红, 施达仁, 梁晓曼, 等. 显色原位杂交和免疫组织化学检测乳腺癌HER-2/neu基因状况和蛋白表达的对照性研究[J]. 中华病理学杂志, 2006, 35(10): 580-583.
[8] 应建明, 郭蕾, 刘秀云, 等. 全自动银增强原位杂交检测乳腺癌患者人表皮生长因子受体2基因状态[J]. 中华医学杂志, 2010, 90(24): 1674-1677.
[9] 许燕, 杨文涛, 杨飞, 等. 双色银染原位杂交与荧光原位杂交在乳腺癌HER-2基因状态评估中的比较[J]. 中华病理学杂志, 2014, 43(4): 226-230.
[10] Wolff, A.C., Hammond, M.E.H., Allison, K.H., et al. (2018) Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Focused Update. Journal of Clinical Oncology, 36, 2105-2122.
https://doi.org/10.1200/JCO.2018.77.8738
[11] Murthy, R.K., Loi, S., Okines, A., et al. (2020) Tucatinib, Trastuzumab, and Capecitabine for HER-2-Positive Metastatic Breast Cancer. The New England Jour-nal of Medicine, 382, 597-609.
https://doi.org/10.1056/NEJMoa1914609
[12] Tarantino, P., Hamilton, E., Tolaney, S.M., et al. (2020) HER-2-Low Breast Cancer: Pathological and Clinical Landscape. Journal of Clinical Oncology, 38, 1951-1962.
https://doi.org/10.1200/JCO.19.02488
[13] 刘月平, 薛卫成, 杨文涛, 步宏. 乳腺癌HER-2低表达病理检测进展及挑战[J]. 中华病理学杂志, 2022, 51(9): 799-802.
[14] 《乳腺癌HER-2检测指南(2019版)》编写组. 乳腺癌HER-2检测指南(2019版) [J]. 中华病理学杂志, 2019, 48(3): 169-175.
[15] Schalper, K.A., Kumar, S., Hui, P., et al. (2014) A Retrospective Population-Based Comparison of HER-2 Immunohistochemistry and Fluorescence in Situ Hybridization in Breast Carcinomas: Impact of 2007 American Society of Clinical Oncology/College of American Pathologists Criteria. Archives of Pathology & Laboratory Medicine, 138, 213-219.
https://doi.org/10.5858/arpa.2012-0617-OA
[16] Fernandez, A.I., Liu, M., Bellizzi, A., et al. (2022) Examination of Low ERBB2 Protein Expression in Breast Cancer Tissue. JAMA Oncology, 8, 607-614.
https://doi.org/10.1001/jamaoncol.2021.7239
[17] Schettini, F., Chic, N., Brasó-Maristany, F., et al. (2021) Clini-cal, Pathological, and PAM50 Gene Expression Features of HER-2-Low Breast Cancer. NPJ Breast Cancer, 7, 1.
https://doi.org/10.1038/s41523-020-00208-2
[18] Lim, S.K., Lee, M.H., Park, I.H., et al. (2016) Impact of Molec-ular Subtype Conversion of Breast Cancers after Neoadjuvant Chemotherapy on Clinical Outcome. Cancer Research and Treatment, 48, 133-141.
https://doi.org/10.4143/crt.2014.262
[19] 白冰, 张艾佳, 郭新, 等. HER2低表达乳腺癌的临床病理特征和预后因素分析[J]. 中国医科大学学报, 2023, 52(6): 494-498.
[20] Prat, A., Bardia, A., Curigliano, G., et al. (2022) An Overview of Clinical Development of Agents for Metastatic or Advanced Breast Cancer without ERBB2 Amplification (HER2-Low). JAMA Oncology.
https://doi.org/10.1001/jamaoncol.2022.4175
[21] Denkert, C., Seither, F., Schneeweiss, A., et al. (2021) Clinical and Molecular Characteristics of HER-2-Low-Positive Breast Cancer: Pooled Analysis of Individual Patient Data from Four Prospective, Neoadjuvant Clinical Trials. The Lancet Oncology, 22, 1151-1161.
https://doi.org/10.1016/S1470-2045(21)00301-6
[22] Zhang, H., Katerji, H., Turner, B.M., et al. (2022) HER-2-Low Breast Cancers: Incidence, HER-2 Staining Patterns, Clinicopathologic Features, MammaPrint and Blue-Print Genomic Profiles. Modern Pathology, 35, 1075-1082.
https://doi.org/10.1038/s41379-022-01019-5
[23] Alves, F.R., Gil, L., Vasconcelos de Matos, L., et al. (2022) Im-pact of Human Epidermal Growth Factor Receptor 2 (HER-2) Low Status in Response to Neoadjuvant Chemotherapy in Early Breast Cancer. Cureus, 14, e22330.
https://doi.org/10.7759/cureus.22330
[24] Li, Y., Abudureheiyimu, N., Mo, H., et al. (2022) In Real Life, Low-Level HER-2 Expression May Be Associated with Better Outcome in HER-2-Negative Breast Cancer: A Study of the National Cancer Center, China. Frontiers in Oncology, 11, Article ID: 774577.
https://doi.org/10.3389/fonc.2021.774577
[25] Gilcrease, M.Z., Woodward, W.A., Nicolas, M.M., et al. (2009) Even Low-Level HER-2 Expression May Be Associated with Worse Outcome in Node-Positive Breast Cancer. The American Journal of Surgical Pathology, 33, 759- 767.
https://doi.org/10.1097/PAS.0b013e31819437f9
[26] Agostinetto, E., Rediti, M., Fimereli, D., et al. (2021) HER-2-Low Breast Cancer: Molecular Characteristics and Prognosis. Cancers (Basel), 13, Article No. 2824.
https://doi.org/10.3390/cancers13112824
[27] Fehrenbacher, L., Cecchini, R.S., Geyer, C.E., et al. (2020) NSABP B-47/NRG Oncology Phase III Randomized Trial Comparing Adjuvant Chemotherapy with or without Trastuzumab in High-Risk Invasive Breast Cancer Negative for HER-2 by FISH and with IHC 1+ or 2. Journal of Clinical Oncology, 38, 444-453.
https://doi.org/10.1200/JCO.19.01455
[28] Ergun, Y., Ucar, G. and Akagunduz, B. (2023) Comparison of HER-2-Zero and HER-2-Low in Terms of Clinicopathological Factors and Survival in Early-Stage Breast Cancer: A Systematic Review and Meta-Analysis. Cancer Treatment Reviews, 115, Article ID: 102538.
https://doi.org/10.1016/j.ctrv.2023.102538
[29] Rossi, V., Sarotto, I., Maggiorotto, F., et al. (2012) Moderate Im-munohistochemical Expression of HER-2 (2+) without HER-2 Gene Amplification Is a Negative Prognostic Factor in Early Breast Cancer. Oncologist, 17, 1418-1425.
https://doi.org/10.1634/theoncologist.2012-0194
[30] Wang, J., Xu, B., Yuan, P., et al. (2014) HER-2 as a Predic-tive Factor for Successful Neoadjuvant Anthracycline Chemotherapy of Locally Advanced and Early Breast Cancer. The International Journal of Biological Markers, 29, e187- e192.
https://doi.org/10.5301/jbm.5000094
[31] Pinhel, I., Hills, M., Drury, S., Salter, J., et al. (2012) ER and HER-2 Expression Are Positively Correlated in HER-2 Non-Overexpressing Breast Cancer. Breast Cancer Research, 14, R46.
https://doi.org/10.1186/bcr3145
[32] Zhang, G., Ren, C., Li, C., et al. (2022) Distinct Clinical and Somatic Muta-tional Features of Breast Tumors with High-, Low-, or Non-Expressing Human Epidermal Growth Factor Receptor 2 Status. BMC Medicine, 20, Article No. 142.
https://doi.org/10.1186/s12916-022-02346-9
[33] Horisawa, N., Adachi, Y., Takatsuka, D., et al. (2022) The Frequency of Low HER-2 Expression in Breast Cancer and a Comparison of Prognosis between Patients with HER-2-Low and HER-2-Negative Breast Cancer by HR Status. Breast Cancer, 29, 234-241.
https://doi.org/10.1007/s12282-021-01303-3
[34] Pegram, M., Hsu, S., Lewis, G., et al. (1999) Inhibito-ry Effects of Combinations of HER-2/Neu Antibody and Chemotherapeutic Agents Used for Treatment of Human Breast Cancers. Oncogene, 18, 2241-2251.
https://doi.org/10.1038/sj.onc.1202526
[35] Friess, T.S. and Bauer, A.M.B. (2002) In Vivo Activity of Recombi-nant Humanized Monoclonal Antibody 2C4 in Xenografts Is Independent of Tumor Type and Degree of HER-2 Over-expression. European Journal of Cancer, 38, S149.
https://doi.org/10.1016/S0959-8049(02)81150-9
[36] Gianni, L., Lladó, A., Bianchi, G., et al. (2010) Open-Label, Phase II, Multicenter, Randomized Study of the Efficacy and Safety of Two Dose Levels of Pertuzumab, a Human Epi-dermal Growth Factor Receptor 2 Dimerization Inhibitor, in Patients with Human Epidermal Growth Factor Receptor 2-Negative Metastatic Breast Cancer. Journal of Clinical Oncology, 28, 1131-1137.
https://doi.org/10.1200/JCO.2009.24.1661
[37] Beck, A., Goetsch, L., Dumontet, C., et al. (2017) Strategies and Challenges for the Next Generation of Antibody-Drug Conjugates. Nature Reviews Drug Discovery, 16, 315-337.
https://doi.org/10.1038/nrd.2016.268
[38] Ferraro, E., Drago, J.Z. and Modi, S. (2021) Implementing Anti-body-Drug Conjugates (ADCs) in HER-2-Positive Breast Cancer: State of the Art and Future Directions. Breast Cancer Research, 23, Article No. 84.
https://doi.org/10.1186/s13058-021-01459-y
[39] Burris, H.A., Tibbitts, J., Holden, S.N., et al. (2011) Trastuzumab Emtansine (T-DM1): A Novel Agent for Targeting HER-2+ Breast Cancer. Clinical Breast Cancer, 11, 275-282.
https://doi.org/10.1016/j.clbc.2011.03.018
[40] Verma, S., Miles, D., Gianni, L., et al. (2012) Trastuzumab Emtansine for HER-2-Positive Advanced Breast Cancer. The New England Journal of Medicine, 367, 1783-1791.
https://doi.org/10.1056/NEJMoa1209124
[41] Yver, A., Agatsuma, T. and Soria, J.C. (2020) The Art of Innovation: Clinical Development of Trastuzumab Deruxtecan and Redefining How Antibody-Drug Conjugates Target HER-2-Positive Cancers. Annals of Oncology, 31, 430- 434.
https://doi.org/10.1016/j.annonc.2019.11.019
[42] Sahin, U., Hartmann, F., Senter, P., Pohl, C., Engert, A., Diehl, V. and Pfreundschuh, M. (1990) Specific Activation of the Prodrug Mitomycin Phosphate by a Bispecific An-ti-CD30/Anti-Alkaline Phosphatase Monoclonal Antibody. Cancer Research, 50, 6944-6948.
[43] Tadokoro, J., Kakihata, K., Shimazaki, M., et al. (2011) Post-Marketing Surveillance (PMS) of All Patients Treated with Irinotecan in Japan: Clinical Experience and ADR Profile of 13,935 Patients. Japanese Journal of Clinical Oncology, 41, 1101-1111.
https://doi.org/10.1093/jjco/hyr105
[44] van der Lee, M.M., Groothuis, P.G., Ubink, R., et al. (2015) The Preclini-cal Profile of the Duocarmycin-Based HER-2-Targeting ADC SYD985 Predicts for Clinical Benefit in Low HER-2-Expressing Breast Cancers. Molecular Cancer Therapeutics, 14, 692-703.
https://doi.org/10.1158/1535-7163.MCT-14-0881-T
[45] Zhu, Y., Zhu, X., Wei, X., et al. (2021) HER-2-Targeted Therapies in Gastric Cancer. Biochimica et Biophysica Acta (BBA)—Reviews on Cancer, 1876, Article ID: 188549.
https://doi.org/10.1016/j.bbcan.2021.188549
[46] Wang, J.Y., et al. (2021) RC48-ADC, a HER-2-Targeting Anti-body-Drug Conjugate, in Patients with HER-2-Positive and HER-2-Low Expressing Advanced or Metastatic Breast Cancer: A Pooled Analysis of Two Studies. Journal of Clinical Oncology, 39, 1022-1022.
https://doi.org/10.1200/JCO.2021.39.15_suppl.1022
[47] Shi, F., Liu, Y., Zhou, X., et al. (2022) Disitamab Ve-dotin: A Novel Antibody-Drug Conjugates for Cancer Therapy. Drug Delivery, 29, 1335-1344.
https://doi.org/10.1080/10717544.2022.2069883
[48] Le Joncour, V., Martins, A., Puhka, M., et al. (2019) A Novel Anti-HER-2 Antibody-Drug Conjugate XMT-1522 for HER-2-Positive Breast and Gastric Cancers Resistant to Trastuzumab Emtansine. Molecular Cancer Therapeutics, 18, 1721-1730.
https://doi.org/10.1158/1535-7163.MCT-19-0207
[49] Hamilton, E.P., et al. (2018) Phase 1 Dose Escalation of XMT-1522, a Novel HER-2-Targeting Antibody-Drug Conjugate (ADC), in Patients (pts) with HER-2-Expressing Breast, Lung and Gastric Tumors. Journal of Clinical Oncology, 36, 2546-2546.
[50] Modi, S., et al. (2019) A Phase III, Multicenter, Randomized, Open Label Trial of [fam-] Trastuzumab Deruxtecan (DS-8201a) versus Investigator’s Choice in HER-2-Low Breast Cancer. Journal of Clinical Oncology, 37, TPS1102.
https://doi.org/10.1200/JCO.2019.37.15_suppl.TPS1102
[51] Bardia, A., et al. (2021) Trastuzumab Deruxtecan (T-DXd, DS-8201) vs Investigator’s Choice of Chemotherapy in Patients with Hormone Receptor-Positive (HR+), HER-2 Low Metastatic Breast Cancer Whose Disease Has Progressed on Endocrine Therapy in the Metastatic Setting: A Randomized, Global Phase 3 Trial (DESTINY-Breast06). Cancer Research, 81, OT-03-09.
https://doi.org/10.1158/1538-7445.SABCS20-OT-03-09
[52] 朱艺, 肖斌, 刘嘉慧, 等. Circ-0003910在HER-2阳性乳腺癌中的表达、定位、生物学作用及蛋白质组学研究[J]. 中国癌症杂志, 2022, 32(10): 979-989.
https://doi.org/10.19401/j.cnki.1007-3639.2022.10.006
[53] Fehrenbacher, L., Cecchini, R.S., Geyer, C.E., et al. (2019) NSABP B-47/NRG Oncology Phase II Randomized Trial Comparing Adjuvant Chemotherapy with or without Trastuzumab in High-Risk Invasive Breast Cancer Negative for HER-2 by FISH and with IHC 1+ or 2.
[54] Xu, K., Bayani, J., Mallon, E., et al. (2022) Discordance between Immunohistochemistry and Erb-B2 Receptor Tyrosine Kinase 2 mRNA to Determine Human Epidermal Growth Factor Receptor 2 Low Status for Breast Cancer. The Journal of Molec-ular Diagnostics, 24, 775-783.
https://doi.org/10.1016/j.jmoldx.2022.04.002
[55] Hicks, D.G., Buscaglia, B., Goda, H., et al. (2018) A Novel Detection Methodology for HER-2 Protein Quantitation in Formalin-Fixed, Paraffin Embedded Clinical Samples Using Fluorescent Nanoparticles: An Analytical and Clinical Validation Study. BMC Can-cer, 18, Article No. 1266.
https://doi.org/10.1186/s12885-018-5172-1
[56] Harigopal, M., Barlow, W.E., Tedeschi, G., et al. (2010) Multiplexed Assessment of the Southwest Oncology Group-Directed Intergroup Breast Cancer Trial S9313 by AQUA Shows That both High and Low Levels of HER-2 Are Associated with Poor Outcome. The American Journal of Pathology, 176, 1639-1647.
https://doi.org/10.2353/ajpath.2010.090711
[57] Won, H.S., Ahn, J., Kim, Y., et al. (2022) Clinical Significance of HER-2-Low Expression in Early Breast Cancer: A Nationwide Study from the Korean Breast Cancer Society. Breast Cancer Research, 24, Article No. 22.
https://doi.org/10.1186/s13058-022-01519-x