您当前的位置:首页 > 学科资讯 > 详情
抗体药物偶联物在人表皮生长因子受体2表达乳腺癌治疗中的进展
浏览量:169     发布者:肿瘤界     时间:2025-07-17


2-1.png


引用本文:李巧佴,黎立喜,李凤娟,等. 抗体药物偶联物在人表皮生长因子受体2表达乳腺癌治疗中的进展[J]. 中国医学前沿杂志(电子版), 2025,17(04):12-18.


作者:李巧佴,黎立喜,李凤娟,王佳玉,马飞 (国家癌症中心 国家临床医学研究中心 中国医学科学院北京协和医学院肿瘤医院肿瘤内科,北京100021)

通信作者:马飞 E-mail:drmafei@126.com


摘要:抗体药物偶联物(antibody drug conjugates,ADCs)是一类新型的抗肿瘤药物,兼具有靶向药物的特异性和化疗药物的高度抗肿瘤活性,极具临床应用前景。近年来,乳腺癌的治疗正进入ADCs治疗时代,以恩美曲妥珠单抗、德曲妥珠单抗等为代表的ADCs,为人表皮生长因子受体2(human epidermal growth factor receptor 2,HER2)阳性乳腺癌患者的治疗带来了新的突破。此外,ADCs的出现,引入了"HER2低表达(HER2-low)"的概念,并为HER2-low的乳腺癌患者带来了生存获益。本综述重点探讨了ADCs在治疗HER2表达乳腺癌中的应用进展。


关键词:乳腺癌;人表皮生长因子受体2阳性;人表皮生长因子受体2低表达;抗体药物偶联物

人表皮生长因子受体2(human epidermal growth factor receptor 2,HER2)阳性是乳腺癌的不良预后因素,HER2阳性(HER2+)患者具有较高的侵袭性和复发率[1]。随着各种靶向HER2药物的应用,显著降低了HER2+乳腺癌复发、转移的风险[2]。抗体药物偶联物(antibody-drug conjugate,ADCs)是一种新型的抗肿瘤药物,它通过将单克隆抗体与细胞毒性药物结合的方式,精准地将毒性药物传递到肿瘤细胞,一方面能增强对肿瘤细胞的杀伤作用,另一方面还可减少对正常细胞的损害[3]。ADCs提升了治疗的针对性,同时也显著增加了治疗效果。近年来,乳腺癌的治疗正在进入ADCs时代,ADCs不仅为HER2+的乳腺癌患者治疗带来了巨大的突破[4],也为部分HER2低表达(HER2-low)的患者带来了生存获益[5],为HER2表达的乳腺癌的治疗迎来转折。本文将综述ADCs在HER2表达乳腺癌,包括HER2+及HER2-low乳腺癌中的进展。



抗体药物偶联物的作用机制


ADCs由单克隆抗体、细胞毒性有效荷载以及连接体3种核心成分构成,同时兼具了抗体的靶点选择性以及细胞毒性药物高杀伤力的特点。ADCs在进入体内后,通过具有高度特异性的单克隆抗体与肿瘤细胞表面靶抗原的结合,可精准地定位到肿瘤细胞并被并其内吞,在细胞内溶酶体的作用下,细胞毒性有效荷载释放,发挥抗肿瘤效应。细胞毒性有效载荷是ADCs的主要效应成分[6,7,8,9]。目前,ADCs的主要细胞毒性有效载荷包括微管蛋白抑制剂(如auristatin类似物和美坦辛类似物)、DNA损伤化合物(如duocarmazine、calicheamicins和pyrrolobenzodiazepines)、RNA聚合酶Ⅱ抑制剂(amanitin)和拓扑异构酶Ⅰ抑制剂(如deruxtecan、govitecan)。同时,单克隆抗体本身也具有一定抗肿瘤作用,如激活补体复合体、激活免疫细胞、诱导抗体依赖性、补体依赖性细胞毒作用,以及激活抗体依赖性细胞介导性吞噬等。


此外,部分ADCs还具有"旁观者效应"。"旁观者"细胞是靶细胞周围的细胞,通常不表达靶抗原的肿瘤细胞。部分ADCs在与靶抗原结合后,有效荷载从靶细胞释放或在靶细胞外释放,进而进入并杀死"旁观者"细胞[9,10]。然而,并非所有的ADCs都具有"旁观者效应"。目前研究认为,ADCs是否具有旁观者效应可能与以下因素有关:①ADCs与靶抗原结合后的内化程度;②连接子性质;③有效荷载的疏水性等。



抗体药物偶联物在人表皮生长因子受2阳性乳腺癌中的应用


现有研究表明,乳腺癌患者中HER2扩增或过表达(免疫组化检测3+或免疫组化检测2+且荧光原位杂交阳性)的发生率为15%~20%[11]。与其他类型乳腺癌相比,该分型患者的生存率较低,复发转移率较高[12,13]。抗HER2药物的出现和发展,使HER2+患者预后得以改善[2],但仍然存在原发或继发耐药的难题。近年来,抗HER2药物的研发取得了显著进展,尤其是以恩美曲妥珠单抗(trastuzumab emtansine,T-DM1)、德曲妥珠单抗(trastuzumab deruxtecan,T-DXd)等为代表的抗HER2 ADCs,其良好的临床疗效及安全性,显示出在治疗HER2+乳腺癌方面的优越性[14,15,16],为HER2+患者带来了新的希望。


2.1 恩美曲妥珠单抗


T-DM1由曲妥珠单抗通过不可裂解的连接子与强效的微管蛋白抑制剂美坦辛(DM1)相连而形成,既保留了曲妥珠单抗的靶向抗肿瘤功能,也保留了DM1的细胞毒作用。T-DM1是首个被美国食品药品监督管理局批准用于治疗HER2+乳腺癌的抗HER2 ADCs,目前已被批准用于新辅助治疗后未达到病理完全缓解的HER2+乳腺癌患者的术后辅助治疗和晚期HER2+乳腺癌患者的挽救治疗[17]


EMILIA研究是一项随机、非盲的Ⅲ期研究,该研究对比了T-DM1与卡培他滨联合拉帕替尼在既往曲妥珠单抗联合紫杉烷治疗失败的HER2+乳腺癌患者中的疗效(n=911),是T-DM1的全球关键性研究。结果显示,T-DM1组与对照组相比,无进展生存期(progression-free survival,PFS)和总生存期(overall survival,OS)均有明显改善(PFS:9.6个月比6.4个月,HR=0.65,P<0.001;OS:29.9个月比25.1个月,HR=0.75,P<0.001),中位随访时间为47.8个月。基于EMILIA研究的结果,美国国家综合癌症网络(National Comprehensive Cancer Network,NCCN)指南推荐T-DM1作为曲妥珠单抗治疗失败后HER2+乳腺癌二线治疗的首选方案。2021年ESMO EMILIA亚洲人群数据显示亚洲患者使用T-DM1作为二线治疗的PFS和OS获益与全球人群结果基本一致,且并未发现新的安全性问题。基于上述结果,2021年国家药品监督管理总局正式批准T-DM1在中国上市。


MARIANNE研究是一线T-DM1+/-帕妥珠单抗对比曲妥珠单抗联合紫杉类(HT)治疗HER2+晚期乳腺癌的随机Ⅲ期临床研究,该研究中含T-DM1方案显示出与HT方案非劣效的疗效,但未达到优效性(objective response rate,ORR):T-DM1+安慰剂组59.7%,T-DM1+帕妥珠单抗组64.2%,HT组67.9%;PFS T-DM1+安慰剂组14.1个月,T-DM1+帕妥珠单抗组15.2个月,HT组13.7个月,≥3级不良反应发生率更低(T-DM1+安慰剂组45.4%;T-DM1+帕妥珠单抗组46.2%;HT组54.1%)[18],具有更好的耐受性。基于该研究,2016版NCCN指南将T-DM1作为HER2+转移性乳腺癌一线治疗的可选方案之一,尤其是在需要减少化疗毒性和改善生活质量的情况下。


在早期乳腺癌的治疗中,T-DM1已成为目前抗HER2新辅助治疗后仍有残存病灶的乳腺癌患者强化辅助治疗的标准治疗方案之一。KATHERINE研究是一项Ⅲ期临床研究,该研究中T-DM1作为新辅助治疗后未达到病理学完全缓解(non-pathological complete response,non-pCR)患者的辅助治疗方案,对其疗效进行了探讨[19]。KATHERINE研究结果显示,对比曲妥珠单抗,T-DM1可显著改善新辅助治疗后non-pCR的HER2+乳腺癌患者的OS率。中位随访8.4年时,患者的7年无浸润性疾病生存率(invasive disease free survival,IDFS)升高(80.8%比67.1%,P<0.0001),OS率亦升高(89.1%比84.4%,HR=0.66,P=0.0027),T-DM1使患者的死亡风险降低34%[20]


然而,在新辅助治疗阶段,KRISTINE研究(Ⅲ期)显示,T-DM1+帕妥珠单抗治疗的病理学完全缓解(pathological complete response,pCR)率低于曲妥珠单抗+帕妥珠单抗+多西他赛+卡铂(44.4%比55.7%,绝对差值为-11.3%,95%CI为20.5~2.0,P=0.016)[21]。以上结果提示,目前化疗联合双靶仍是HER2+乳腺癌患者新辅助治疗的首选。


现有研究显示,T-DM1安全性可控。既往各研究中T-DM1的安全性数据基本一致,常见的1~2级不良事件为胃肠道毒性、神经毒性和左室射血分数下降;常见的3~4级不良事件是血小板减少以及转氨酶水平升高,其中3~4级的血小板减少是T-DM1的主要剂量限制性毒性反应,也是亚洲人群常见的不良反应[17,22,23]


2.2 德曲妥珠单抗


T-DXd是第2个获批的抗HER2 ADCs。与T-DM1类似,T-DXd由曲妥珠单抗通过1种可裂解连接子与拓扑异构酶Ⅰ抑制剂依沙替康衍生物组成[24]。与T-DM1不同的是,T-DXd的连接子可被靶细胞中的溶酶体酶裂解,且依沙替康衍生物具有膜通透性[25,26],这使得T-DXd能够发挥旁观者效应,克服HER2的异质性,从而更有效地杀伤肿瘤细胞。T-DXd已被批准用于治疗既往接受过1种或1种以上抗HER2药物治疗的不可切除或转移性HER2+乳腺癌患者[27]


DESTINY-BREAST03研究(Ⅲ期,前瞻性)中头对头对比了T-DXd和T-DM1在既往接受过曲妥珠单抗或者TH药物治疗的HER2+晚期乳腺癌患者中的疗效及安全性(n=524)[27]。该研究结果显示,T-DXd的疗效显著优于T-DM1[27,28]。T-DXd相对于T-DM1 PFS绝对值获益提高了近2年(25.1个月比7.2个月,HR=0.33,P<0.000001)。在疾病缓解方面,T-DXd也展现出了明显的优势,T-DXd和T-DM1的ORR分别为79.7%、34.2%(绝对差值45.5%,95%CI为37.6~53.4)。基于上述结果,T-DXd已被批准用于HER2+晚期乳腺癌患者的二线治疗。2024年美国临床肿瘤学会(American Society of Clinical Oncology,ASCO)更新了DESTINY-BREAST03研究的中位PFS、OS数据[29],分别为29个月和52.6个月,进一步巩固了德曲妥珠单抗在HER2+晚期乳腺癌二线治疗中的标准地位。


DESTINY-BREAST07是一项Ⅰb/Ⅱ期多中心、开放标签、多队列研究,旨在探索T-DXd+/-帕妥珠单抗一线治疗HER2+转移性乳腺癌(metastatic breast cancer,mBC)的安全性、耐受性和抗肿瘤活性[30]。2024年ASCO公布了该研究中期分析结果[30],发现无论是否联合帕妥珠单抗,T-DXd均显示出优异的抗肿瘤活性,其ORR均达到80%左右,12个月PFS均>80%,而且无论是单药组还是联合组,均未发现新的不良反应事件。该研究为HER2+乳腺癌的一线治疗提供了新的治疗选择和思路,为后续的Ⅲ期临床研究(DESTINY-BREAST09)提供了有力的基础和支持,有望重新定义HER2 +晚期乳腺癌一线治疗的新标准,进一步延长患者的生存。


此外,T-DXd在HER2+乳腺癌脑转移患者的治疗中也显示出良好的疗效。DESTINY-BREAST12是一项聚焦于HER2+乳腺癌脑转移治疗的Ⅲb/Ⅳ期的前瞻性研究[31]。2024年欧洲肿瘤内科学会(European Society for Medical Oncology,ESMO)首次公布了DESTINY-BREAST12研究[32]的结果。该研究结果显示,T-DXd治疗HER2+乳腺癌脑转移患者的中位PFS为17.3个月,进一步验证了T-DXd在HER2+晚期乳腺癌治疗中的良好疗效。


目前,T-DXd在HER2+早期乳腺癌患者的辅助强化及新辅治疗的相关研究也进行了相应的布局,如DESTINY-BREAST05研究头对头对比了T-DXd和T-DM1用于治疗接受新辅助治疗后non-PCR的高风险HER2+乳腺癌患者的安全性和有效性;而DESTINY-BREAST11研究,则旨在探索T-DXd单药或序贯紫杉醇联合曲妥珠单抗、帕妥珠单抗对比标准治疗用于HER2+乳腺癌患者新辅助治疗的获益情况。


在既往各项临床研究中,T-DXd的安全性均具有可控性。与T-DXd相关的毒性作用以1~2级的消化道反应和骨髓抑制最为常见,而常见的高级别不良事件是中性粒细胞计数减少和贫血。消化道反应主要表现为恶心、呕吐等,DESTINY-BREAST01研究中两者的发生率分别为77.7%和45.7%[33]。NCCN指南已推荐按照高致吐风险管理措施进行T-DXd治疗前的预防性止吐。


此外,一些接受T-DXd治疗的患者出现了不同严重程度的间质性肺疾病[27,34,35]。值得关注的是,相较于T-DM1,T-DXd组患者不良反应发生率更高,T-DXd与T-DM1药物相关不良事件发生率分别为98.1%和86.6%,3级或4级药物相关不良事件发生率分别为45.1%和39.8%[27]


2.3 维迪西妥单抗


维迪西妥单抗(disitamabvedotin,RC48)是我国自主研发的第一个抗HER2 ADCs,是由一种新的抗HER2单抗(disitamab)通过可切割连接子与小分子微管蛋白抑制剂甲基澳瑞他汀E(MMAE)偶联而成[36],对表达HER2的肿瘤细胞具有强杀伤作用,同时还具有旁观者效应。2021年ASMO会议报道了RC48治疗HER2+乳腺癌患者的ORR为42.9%,DCR率为90.5%,中位PFS为6.6个月[37]。RC48已经在国内上市,是国内首个获批上市的ADCs。目前,RC48治疗HER2+肝转移乳腺癌患者的Ⅲ期临床试验(RC48-C006)已达主要研究终点。2024年圣安东尼奥乳腺癌研讨会(San Antonio Breast Cancer Symposium,SABCS)报道了该研究的结果,与拉帕替尼联合卡培他滨相比,RC48可显著延长患者的PFS(9.9个月比4.9个月,HR=0.561, P=0.0143)有望进一步丰富乳腺癌患者的治疗选择。



抗体药物偶联物在人表皮生长因子受体2低表达乳腺癌中的应用


HER2-low(免疫组化检测1+或免疫组化检测2+且荧光原位杂交阴性)[38]乳腺癌患者约占所有乳腺癌的1/2[39],传统抗HER2靶向药物对HER2-low乳腺癌患者的疗效十分有限[40,41,42]。激素受体(hormone receptor,HR)阴性/HER2-low乳腺癌既往被归入三阴性乳腺癌(triple negative breast cancer,TNBC),治疗以化疗、免疫治疗为主;而HR阳性/HER2-low则被归入Luminal型乳腺癌,晚期患者的治疗以内分泌治疗或内分泌联合靶向治疗为主。随着ADCs的出现,HER2-low乳腺癌的治疗模式也发生了显著改变[43]。有研究发现,即使不依赖于HER2通路,HER2-low患者也可能从新型抗HER2 ADCs治疗中获益。目前NCCN、ESMO等国际权威指南已将HER2-Low作为新的治疗分型。


3.1 抗人表皮生长因子受体2抗体药物偶联物


DS-8201-A-J101研究首次证实了T-DXd在HER2-low乳腺癌患者中的抗肿瘤活性,并由此拉开了HER2-low乳腺癌临床研究的序幕[44]。基于该研究,2021版《HER2阳性乳腺癌临床诊疗专家共识》也首次引入了"HER2-low"的概念[43]。DESTINY-BREAST04研究[45]是首个针对HER2-low亚型的Ⅲ期临床研究,旨在评估T-DXd治疗HER2-low转移性乳腺癌的疗效及安全性。该研究结果显示,在既往接受过1~2线化疗(HR阳性患者需接受过≥1线内分泌治疗后进展且不再从内分泌治疗中获益)的HER2-low晚期乳腺癌患者中,相较于医生选择化疗(treatment of physician′s choice,TPC),T-DXd可显著改善患者的PFS和OS(中位PFS:9.9个月比5.4个月,HR=0.51, P<0.001;中位OS:23.4个月比16.8个月,HR=0.64,P=0.001),证实T-DXd治疗可为HER2-low晚期乳腺癌患者带来生存获益。同时,基于此结果,T-DXd于2022年获批用于HER2-low转移性乳腺癌的治疗。


此外,DESTINY-BREAST06研究将T-DXd前移至HR阳性/HER2-low患者的一线治疗,2024年SABCS汇报的该研究[46]结果显示,T-DXd可显著改善该亚型患者的PFS(13.2个月比8.1个月,HR=0.632,P<0.0001),提示其可以作为一种治疗选择。TRIO-US B-12 TALENT研究(Ⅱ期)[47]首次将T-DXd用于HR阳性/HER2-low早期乳腺癌的新辅助治疗,该研究目前尚处于临床试验阶段,其结果值得期待。在联合治疗方面,DESTINY-BREAST08研究是一项Ⅰb期、多中心、开放标签研究,旨在评估T-DXd联合卡培他滨或卡匹色替在HER2-low转移性乳腺癌患者中的疗效及安全性。2024年SABCS公布的该研究[48]结果显示,T-DXd联合卡培他滨或联合卡匹色替均显示出良好的抗肿瘤活性(ORR均为60%)。BEGONIA研究的队列6评估了"度伐利尤单抗联合T-DXd"一线治疗HER2-low TNBC的疗效,在前期数据中已观察到令人鼓舞的有效性和可控的安全性[49]


与T-DXd类似,RC48也具有旁观者效应,可有效杀伤HER2-low细胞。一项关于RC48的两项Ⅰ期研究(C001和C003)的汇总分析显示,在HER2-low亚组中(n=48),ORR和PFS分别为39.6%和5.7个月[37]。尽管PFS和ORR未超过T-DXd的Ⅰ期研究数据(ORR 37%,PFS 11.1个月)范围[50](DS-8201-A-J101),但疗效仍优于其他传统治疗方案。这提示RC48在HER2-low晚期乳腺癌中具有良好的应用前景,值得进一步探索。目前,针对HER2-low乳腺癌的Ⅲ期临床研究C012尚在进行中(NCT04400695)。


尽管T-DM1在HER2+乳腺癌患者中显示出优异的疗效,然而回顾性分析发现T-DM1的疗效依赖于HER2的表达水平,在HER2-low乳腺癌亚型中的疗效十分有限[51]


3.2 非人表皮生长因子受体2靶点的抗体药物偶联物


以戈沙妥珠单抗(sacituzumabgovitecan,SG)为代表的非HER2靶点的ADCs(如靶向TROP2、HER3、LIV-1、BH-74等)在TNBC中均取得了不错的疗效[34,52,53,54],SG已成为TNBC二线及以后的治疗选择。HR阴性/HER2-low乳腺癌既往被归于TNBC范畴,这表明HER2-low乳腺癌和TNBC存在一部分重叠人群。有研究显示TNBC中HER2-low人群占比为36.6%[55],因而这些非HER2靶点的ADCs亦可作为HER2-low晚期乳腺癌患者的治疗选择。


T-DXd和SG均已成为HER2-low晚期乳腺癌患者的治疗选择,但二者的排序尚缺乏头对头临床研究验证。基于二者疗效和安全性比较,国内外相关指南一致推荐在治疗HER2-low晚期乳腺癌时,应优先考虑T-DXd,对于免疫组化检测为0或不适用T-DXd的患者,SG亦可作为治疗的选择。



展 望


近年来,ADCs的问世彻底变革了乳腺癌的治疗模式。抗HER2 ADCs凭借其独特的分子结构,利用HER2靶向抗体的特异性,能够将细胞毒性有效载荷精准锚定至HER2过表达的肿瘤细胞,进而发挥强大的抗肿瘤效应。以T-DXd和T-DM1等为代表的抗HER2 ADCs已成为HER2+乳腺癌患者的标准治疗药物,进一步改善了HER2+乳腺癌患者的生存。随着ADCs的出现,HER2-low的概念被引入,HER2-low乳腺癌已成为独立的治疗分型。该亚型的患者不仅能够从抗HER2 ADCs治疗中有生存获益,还有望从其他非HER2靶点的ADCs治疗中受益。同时,ADCs在乳腺癌治疗领域仍面临诸多亟待解决的问题,如生物标志物检测、耐药机制的探索、ADC药物结构优化以及联合治疗的应用等,均是未来临床研究的重要方向。



参考文献


[1]SWAIN SM , SHASTRY M , HAMILTON E .Targeting HER2-positive breast cancer:Advances and future directions[J]. Nat Rev Drug Discov, 2023,22(2):101-216.

 [2]DAWOOD S , BROGLIO K , BUZDAR AU ,et al. Prognosis of women with metastatic breast cancer by HER2 status and trastuzumab treatment:An institutional-based review[J]. J Clin Oncol, 2010,28(1):92-98.

 [3]KHERA E , CILLIERS C , SMITH MD ,et al. Quantifying ADC bystander payload penetration with cellular resolution using pharmacodynamic mapping[J]. Neoplasia, 2021,23(2):210-221.

 [4]ZHU K , YANG X , TAI H ,et al. HER2-targeted therapies in cancer:A systematic review[J]. Biomark Res , 2024,12(1):16.

 [5]CORTI C , GIUGLIANO F , NICOLO E ,et al. HER2-low breast cancer:A new subtype? [J]. Curr Treat Options Oncol, 2023,24(5):468-478.

 [6]RITCHIE M , TCHISTIAKOVA L , SCOTT N .Implications of receptor-mediated endocytosis and intracellular trafficking dynamics in the development of antibody drug conjugates[J]. MAbs, 2013,5(1):13-21.

 [7]GEBLEUX R , STRINGHINI M , CASANOVA R ,et al. Non-internalizing antibody-drug conjugates display potent anti-cancer activity upon proteolytic release of monomethyl auristatin E in the subendothelial extracellular matrix[J]. Int J Cancer, 2017,140(7):1670-1679.

 [8]KALIM M , CHEN J , WANG S ,et al. Intracellular trafficking of new anticancer therapeutics:Antibody-drug conjugates[J]. Drug Des Devel Ther, 2017,11:2265-2276.

 [9]YU J , FANG T , YUN C ,et al. Antibody-drug conjugates targeting the human epidermal growth factor receptor family in cancers[J]. Front Mol Biosci, 2022,9:847835.

 [10]STAUDACHER AH , BROWN MP . Antibody drug conjugates and bystander killing:Is antigen-dependent internalisation required? [J]. Br J Cancer, 2017,117(12):1736-1742.

 [11]NAJJAR MK , MANORE SG , REGUA AT ,et al. Antibody-drug conjugates for the treatment of HER2-positive breast cancer[J]. Genes(Basel), 2022,13(11):2065.

 [12]STANOWICKA-GRADA M , SENKUS E .Anti-HER2 drugs for the treatment of advanced HER2 positive breast cancer[J]. Curr Treat Options Oncol, 2023,24(11):1633-1650.

 [13]HUDIS CA . Trastuzumab——mechanism of action and u se in clinical practice [J]. N Engl J Med, 2007,357(1):39-51.

 [14]SAINI KS , AZIM JR HA , METZGER-FILHO O ,et al. Beyond trastuzumab:New treatment options for HER2-positive breast cancer[J]. Breast, 2011,20:Suppl 3-S20. S27

 [15]NARAYAN P , OSGOOD CL , SINGH H ,et al. FDA approval summary:Fam-trastuzumab deruxtecan-nxki for the treatment of unresectable or metastatic HER2-positive breast cancer[J]. Clin Cancer Res, 2021,27(16):4478-4485.

 [16]WEDAM S , FASHOYIN-AJE L , GAO X ,et al. FDA approval ssummary:Ado-trastuzumab emtansine for the adjuvant ttreatment of HER2-positive early breast cancer[J]. Clin Cancer Res, 2020,26(16):4180-4185.

 [17]VON MINCKWITZ G , HUANG CS , MANO MS ,et al. Trastuzumab emtansine for residual invasive HER2-positive breast cancer[J]. N Engl J Med, 2019,380(7):617-628.

 [18]PEREZ EA , BARRIOS C , EIERMANN W ,et al. Trastuzumab emtansine with or without pertuzumab versus trastuzumab plus taxane for hhuman epidermal growth factor receptor 2-positive,advanced breast cancer:Primary results from the phase Ⅲ MARIANNE study[J]. J Clin Oncol, 2017,35(2):141-148.

 [19]HUANG CS , YANG Y , KWONG A ,et al. Trastuzumab emtansine(T-DM1)versus trastuzumab in Chinese patients with residual invasive disease after neoadjuvant chemotherapy and HER2-targeted therapy for HER2-positive breast cancer in the phase 3 KATHERINE study[J]. Breast Cancer Res Treat, 2021,187(3):759-768.

 [20]LOIBL S , MANO MS , UNTCH M ,et al. Abstract GS03-12:Phase Ⅲ study of adjuvant ado-trastuzumab emtansine vs trastuzumab for residual invasive HER2-positive early breast cancer after neoadjuvant chemotherapy and HER2-targeted therapy:KATHERINE final IDFS and updated OS analysis[J]. Cancer Res, 2024,84(9_Supplement):GS03-12.

 [21]HURVITZ SA , MARTIN M , SYMMANS WF ,et al. Neoadjuvant trastuzumab,pertuzumab,and chemotherapy versus trastuzumab emtansine plus pertuzumab in patients with HER2-positive breast cancer(KRISTINE):A randomised,open-label,multicentre,phase 3 trial[J]. Lancet Oncol, 2018,19(1):115-126.

 [22]WELSLAU M , DIERAS V , SOHN JH ,et al. Patient-reported outcomes from EMILIA,a randomized phase 3 study of trastuzumab emtansine(T-DM1)versus capecitabine and lapatinib in human epidermal growth factor receptor 2-positive locally advanced or metastatic breast cancer[J]. Cancer, 2014,120(5):642-651.

 [23]KROP IE , KIM SB , GONZALEZ-MARTIN A ,et al. Trastuzumab emtansine versus treatment of physician ' s choice for pretreated HER2-positive a dvanced breast cancer(TH3RESA):A randomised,open-label,phase 3 trial [J]. Lancet Oncol, 2014,15(7):689-699.

 [24]OGITANI Y , AIDA T , HAGIHARA K ,et al. DS-8201a,anovel HER2-targeting ADC with a novel dnatopoisomerase iinhibitor,demonstrates a promising antitumor efficacy with differentiation from T-DM1[J]. Clin Cancer Res, 2016,22(20):5097-5108.

 [25]FERRARO E , DRAGO JZ , MODI S .Implementing antibody-drug conjugates(ADCs)in HER2-positive breast cancer:State of the art and future directions[J]. Breast Cancer Res, 2021,23(1):84.

 [26]HA SYY , ANAMI Y , YAMAZAKI CM ,et al. An enzymatically cleavable tripeptide linker for maximizing the therapeutic index of antibody-drug conjugates[J]. Mol Cancer Ther, 2022,21(9):1449-1461.

 [27]CORTES J , KIM SB , CHUNG WP ,et al. Trastuzumab deruxtecan versus trastuzumab emtansine for breast cancer[J]. N Engl J Med, 2022,386(12):1143-1154.

 [28]HURVITZ SA , HEGG R , CHUNG WP ,et al. Trastuzumab deruxtecan versus trastuzumab emtansine in patients with HER2-positive metastatic breast cancer:Updated results from DESTINY-Breast03,a randomised,open-label,phase 3 trial[J]. Lancet, 2023,401(10371):105-117.

 [29]HAMILTON EP , HURVITZ SA , IM SA ,et al. Trastuzumab deruxtecan(T-DXd)vs trastuzumab emtansine(T-DM1)in patients(pts)with HER2+ metastatic breast cancer(mBC):Updated survival results of DESTINY-Breast03[J]. J Clin Oncol, 2024,42(16_suppl):1025.

 [30]ANDRE F , HAMILTON EP , LOI S ,et al. DESTINY-Breast07:Dose-expansion interim analysis of T-DXd monotherapy and T-DXd + pertuzumab in patients with previously untreated HER2 + mBC [J]. J Clin Oncol, 2024,42(16_suppl):1009.

 [31]HARBECK N , CIRUELOS E , JERUSALEM G ,et al. Trastuzumab deruxtecan in HER2-positive advanced breast cancer with or without brain metastases:A phase 3b/4 trial[J]. Nat Med, 2024,30(12):3717-3727.

 [32]LIN N , CIRUELOS EM , JERUSALEM G ,et al. LBA18 trastuzumab deruxtecan(T-DXd)in patients(pts)with HER2+ advanced/metastatic breast cancer(mBC)with or without brain metastases(BM):DESTINYBreast-12 primary results[J]. Ann Oncol, 2024,35(2_suppl):S1211-S1212.

 [33]MODI S , SAURA C , YAMASHITA T ,et al. Trastuzumab deruxtecan in previously treated HER2-positive breast cancer[J]. N Engl J Med, 2020,382(7):610-621.

 [34]KROP IE , MASUDA N , MUKOHARA T ,et al. Patritumab deruxtecan(HER3-DXd),a human epidermal growth factor receptor 3-directed antibody-drug conjugate,in patients with previously treated human epidermal growth factor receptor 3-expressing metastatic breast cancer:A multicenter,phase Ⅰ/Ⅱ trial[J]. J Clin Oncol, 2023,41(36):5550-5560.

 [35]MODI S , SAURA C , YAMASHITA T ,et al. Abstract PD3-06:Updated results from DESTINY-breast01,a phase 2 trial of trastuzumab deruxtecan(T-DXd)in HER2 positive metastatic breast cancer[J]. Cancer Res, 2021,81(4_Suppl):PD3-06.

 [36]LI H , YU C , JIANG J ,et al. An anti-HER2 antibody conjugated with monomethyl auristatin E is highly effective in HER2-positive human gastric cancer[J]. Cancer Biol Ther, 2016,17(4):346-354.

 [37]WANG J , LIU Y , ZHANG Q ,et al. RC48-ADC,a HER2-targeting antibody-drug conjugate,in patients with HER2-positive and HER2-low expressing advanced or metastatic breast cancer:A pooled analysis of two studies[J]. J Clin Oncol, 2021,39(15_suppl):1022.

 [38]中国临床肿瘤学会乳腺癌专家委员会中国抗癌协会乳腺癌专业委员会.人表皮生长因子受体2阳性乳腺癌临床诊疗专家共识(2021版)[J]. 中华医学杂志, 2021,101(17):1226-1231.

 [39]WOLFF AC , SOMERFIELD MR , DOWSETT M ,et al. Human epidermal growth factor receptor 2 testing in breast cancer:ASCO-college of american pathologists guideline update[J]. J Clin Oncol, 2023,41(22):3867-3872.

 [40]TARANTINO P , HAMILTON E , TOLANEY SM ,et al. HER2-low breast cancer:Pathological and clinical landscape[J]. J Clin Oncol, 2020,38(17):1951-1962.

 [41]TOZBIKIAN G , KRISHNAMURTHY S , BUI MM ,et al. Emerging landscape of targeted therapy of breast cancers with low human epidermal growth factor receptor 2 protein expression[J]. Arch Pathol Lab Med, 2024,148(2):242-255.

 [42]PRAT A , BARDIA A , CURIGLIANO G ,et al. An overview of clinical development of agents for metastatic or advanced breast cancer without ERBB2 amplification(HER2-low)[J]. JAMA Oncol, 2022. 10.1001/jamaoncol.2022.4175 .

 [43]黄香,蒋梦萍,包胜南,等. 2021年CSCO《乳腺癌诊疗指南》更新要点解读[J]. 中国肿瘤外科杂志, 2021,13(3):209-215.

 [44]DOI T , SHITARA K , NAITO Y ,et al. Safety,pharmacokinetics,and antitumour activity of trastuzumab deruxtecan(DS-8201),a HER2-targeting antibody-drug conjugate,in patients with advanced breast and gastric or gastro-oesophageal tumours:A phase 1 dose-escalation study[J]. Lancet Oncol, 2017,18(11):1512-1522.

 [45]MODI S , JACOT W , YAMASHITA T ,et al. Trastuzumab deruxtecan in previously treated HER2-low advanced breast cancer[J]. N Engl J Med, 2022,387(1):9-20.

 [46]CURIGLIANO G , HU X , DENT RA ,et al. Trastuzumab deruxtecan(T-DXd)vs physician′s choice of chemotherapy(TPC)in patients(pts)with hormone receptor-positive(HR + ),human epidermal growth factor receptor 2(HER2)-low or HER2-ultralow metastatic breast cancer(mBC)with prior endocrine therapy(ET):Primary results from DESTINY-Breast06(DB-06) [J]. J Clin Oncol, 2024,42(17_suppl):LBA1000.

 [47]HURVITZ SA , PEDDI PF , TETEF ML ,et al. TRIO-US B-12 TALENT:Phase Ⅱ neoadjuvant trial evaluating trastuzumab deruxtecan with or without anastrozole for HER2-low,HR + early stage breast cancer [J]. J Clin Oncol, 2021,39(15_suppl):TPS603.

 [48]JHAVERI K , ANDRÉ F , HAMILTON E ,et al. Abstract RF02-03:Trastuzumab deruxtecan(T-DXd)in combination with anastrozole or fulvestrant in patients with HER2-low HR + advanced/metastatic breast cancer:A phase 1b,open-label,multicenter,dose-expansion study(DESTINY-Breast08) [J]. Cancer Res, 2024,84(9_Suppl):RF02-03.

 [49]SCHMID P , IM S , ARMSTRONG A ,et al. BEGONIA:Phase 1b/2 study of durvalumab(D)combinations in locally advanced/metastatic triple-negative breast cancer(TNBC)—initial results from ar m 1,d+paclitaxel(P),and arm 6,d+trastuzumab deruxtecan(T-DXd) [J]. J Clin Oncol, 2021,39:1023.

 [50]MODI S , PARK H , MURTHY RK ,et al. Antitumor activity and safety of trastuzumab deruxtecan in patients with HER2-low-expressing advanced breast cancer:Results from a phase Ⅰb study[J]. J Clin Oncol, 2020,38(17):1887-1896.

 [51]KROP IE , LORUSSO P , MILLER KD ,et al. A phase Ⅱ study of trastuzumab emtansine in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer who were previously treated with trastuzumab,lapatinib,an anthracycline,a taxane,and capecitabine[J]. J Clin Oncol, 2012,30(26):3234-3241.

 [52]CAREY LA , LOIRAT D , PUNIE K ,et al. Sacituzumab govitecan as second-line treatment for metastatic triple-negative breast cancer-phase 3 ASCENT study subanalysis[J]. NPJ Breast Cancer, 2022,8(1):72.

 [53]RIZZO A , CUSMAI A , ACQUAFREDDA S ,et al. Ladiratuzumab vedotin for metastatic triple negative cancer:Preliminary results,key challenges,and clinical potential[J]. Expert Opin Investig Drugs, 2022,31(6):495-498.

 [54]KINNEER K , WORTMANN P , COOPER ZA ,et al. Design and preclinical evaluation of a novel B7-H4-directed antibody-drug conjugate,AZD8205,alone and in combination with the PARP1-sselective inhibitor AZD5305[J]. Cli n Cancer Res , 2023,29(6):1086-1101.

 [55]SCHETTINI F , CHIC N , BRASO-MARISTANY F ,et al. Clinical,pathological,and PAM50 gene expression features of HER2-low breast cancer[J]. NPJ Breast Cancer, 2021,7(1):1.



来源:肿瘤界