您好,欢迎您

【35under35】赖建国医生: HER2低表达乳腺癌迎来里程碑式的精准治疗时代?

2023年08月22日
作者:赖建国
医院:广东省人民医院

 

赖建国
主治医师

广东省人民医院  乳腺科
中山大学孙逸仙纪念医院乳腺外科博士,中共党员,科研秘书,纪检委员
擅长外科为主导的乳腺癌多学科综合治疗
至今已发表SCI论文20余篇,累计总影响因子122.598,包括: EBioMedicine, Advanced Science,Ther Adv Med Oncol, Journal of Translational Medicine, Advanced Therapeutics等国际著名杂志
主持国家自然科学基金面上项目一项
主持国家自然科学基金青年项目一项  
主持广东省人民医院科技专项(人才类)一项
主持广州市基础研究计划基础与应用基础研究项目一项
主持广东省医学科研基金项目一项
参与国家自然科学基金面上项目两项
参与广东省重点领域研发计划项目(新一代人工智能)一项
中国抗癌协会肿瘤标志专业委员会 委员
广东省抗癌协会肿瘤分子医学专业委员会 委员
广东省抗癌协会抗肿瘤药物专业委员会 委员
广东省抗癌协会肿瘤转移委员会委员 委员
广东省基层医药学会乳腺癌专业委员会 委员

基于世界卫生组织国际癌症研究机构的最新数据,2020年全球乳腺癌新发病例高达226万例,超过了肺癌220万例。乳腺癌取代肺癌,成为全球第一大癌症。乳腺癌是女性最常见的恶性肿瘤[1-4]。在我国2020年乳腺癌患者约42万,乳腺癌死亡人数约12万,约占全球乳腺癌死亡人数18% [5]。其中,HER2低表达乳腺癌约占乳腺癌总体人群的45%-55%,亟需更精准的治疗,使患者生存获益[6-8]。笔者结合HER2低表达乳腺癌的最新研究进展和我科真实世界研究进行阐述总结,以飨读者。

1.HER2低表达乳腺癌的定义

人表皮生长因子受体2(human epidermal growth factor receptor 2, HER2,又称ERBB2)是一种酪氨酸激酶受体,编码受体型酪氨酸激酶生长因子跨膜糖蛋白,激活下游酪氨酸激酶级联信号,从而促进乳腺癌细胞的增殖、侵袭和迁移[9-12]。约15-20%乳腺癌发生HER2异常扩增/过表达,抗HER2靶向治疗显著改善了HER2阳性乳腺癌患者的生存预后[13, 14]。根据2018年ASCO与 CAP联合颁布的《乳腺癌HER2检测指南》,HER2分子检测运用阳性或阴性两分法,即HER2阳性:免疫组织化学染色(immunohistochemistry,IHC)评分3+、或2+且荧光原位杂交(fluorescence in situ hybridization,FISH)阳性。HER2阴性:IHC结果为0,+,或2+且FISH阴性[15]。稍显粗暴的HER2表达二分法,使得约一半乳腺癌患者(HER2 1+或HER2 2+且FISH阴性),归为HER2低表达类型,不推荐使用曲妥珠单抗(Trastuzumab)靶向治疗。因此,为实现乳腺癌精准治疗,HER2表达三分法孕育而生,即:HER2零表达、HER2低表达和HER2阳性[16]

图片12.png图1. HER2表达三分法(https://www.sabcs.org/)

2. HER2低表达乳腺癌是一个独立的新亚型吗?

Carsten等学者对2310例乳腺癌患者进行汇总分析发现:相较于HER2零表达乳腺癌,HER2低表达乳腺癌具有更高的HR阳性比例(64.0% vs 36.7%,P<0.0001)、N分期、组织学分级和Ki67具有显著统计学差异。相较于HER2零表达乳腺癌,HER2低表达乳腺癌具有更低的病理完全缓解率(pathological complete response,pCR)(29.2% vs 39.0% , P=0.0002)。相较于HER2零表达乳腺癌,HER2低表达乳腺癌具有更好的无疾病生存(Disease-free survival,DFS, 83.4% vs 76.1%, P=0.0084)和总生存(Overall survival, OS, 91.6% vs 85.8%, P=0.0016)[8]。Schettini等学者的3689例HER2阴性乳腺癌研究表明:相较于HER2零表达乳腺癌患者,HER2低表达乳腺癌患者具有更大的年龄、更高的T分期、更高的N分期、更高的组织学分级、更高的HR阳性比例,而两组人群OS无统计学差异(P=0.234)[17]。Horisawa等学者对4918例乳腺癌患者大样本分析揭示,HER2低表达乳腺癌和HER2零表达乳腺癌患者之间的DFS和OS无显著统计学差异[18]。Peiffer等学者对美国国家癌症数据库1136016例乳腺浸润性癌患者分析发现:相较于HER2零表达乳腺癌,HER2低表达乳腺癌具有更低的pCR率(16.3% vs 23.6%)。

该研究揭示了HER2低表达乳腺癌在HR阳性和三阴性乳腺癌TNBC中更常见。HER2表达和HR表达及组织学状态之间存在关联,但在治疗和生存预后上仅存在轻微差异,这些并不支持将HER2低表达乳腺癌作为一个独特的新亚型。HER2的抗体药物偶联物ADC可显著提高HER2低表达乳腺癌的生存预后,但这并不是由于HER2低表达和HER2零表达乳腺癌之间生物学行为的内在差异所导致的[19]。2022-2023年欧洲肿瘤内科学会(ESMO)邀请欧洲、美洲、亚洲9个国家共32位乳腺癌治疗领域带头专家召开多学科专家共识会议,针对ESMO临床实践指南尚未详细涵盖的HER2低表达乳腺癌,达成HER2低表达乳腺癌ESMO专家共识[16]。该共识指出:

(1).对HR表达进行校正后,HER2低表达与HER2零表达乳腺癌的分子差异不显著。因此,HER2低表达乳腺癌不应该被看作独特的分子亚型,而应被视为具有异质性的肿瘤,其生物学特征主要取决于HR表达。

(2).目前,尚缺乏充足证据将有意义的生存预后归因于HER2低表达。对HR表达进行校正后,大多数研究中并未发现HER2低表达与HER2零表达相比,两组乳腺癌患者生存预后具有显著统计学差异。既往许多研究已经对HER2低表达与HER2零表达乳腺癌的临床病理特征进行分析,但二者之间体细胞突变的差异尚不清楚。针对这一难题,我科廖宁教授发起了我院HER2低表达乳腺癌的真实世界研究,利用523例中国女性人群的520个基因Panel的二代测序(NGS)数据分析,纳入了HER2零表达(N=90例),HER2低表达(N=231例)和HER2阳性(N=202例)[20]

该研究揭示了:在临床特征方面,相较于HER2零表达乳腺癌,HER2低表达患者更倾向于HR阳性,Ki67低表达,新辅助治疗后pCR率更低。我们进一步分析了总体队列中149名接受新辅助治疗患者的治疗结果。HER2低表达亚组pCR率明显更低,仅15.9%,而HER2零表达亚组为37.5%(P=0.042),HER2阳性亚组则高达51.6%(P<0.001)。而在DFS生存预后方面,HER2低表达和零表达两个亚组之间则没有显著性统计差异(P=0.271),且不受病理完全缓解、HR状态或基于IHC何种分子分型的影响。而在基因突变特征方面,与HER2零表达乳腺癌相比,HER2低表达乳腺癌的基因突变谱及信号通路突变方面具有明显的差异,表现为更高的PIK3CA突变率以及更低的TP53突变率。而HER2零表达的乳腺癌则有更多免疫检查点相关基因突变,更多范可尼贫血相关基因突变,及更多的P53信号通路及细胞周期通路的相关基因突变。

图片13.png图2. HER2不同亚组突变基因显著差异(A)和分子通路(B)[20]

3.HER2低表达乳腺癌的治疗进展

(1)单克隆抗体(Monoclonal Antibodies)
Trastuzumab可直接与HER2受体蛋白细胞外侧段的 IV 区进行特异性结合,阻断HER2或与其他 HER 家族成员组成二聚体,从而减少了恶性肿瘤增殖信号传递,是公认的显著改善HER2阳性乳腺癌生存预后的HER2单克隆抗体[10, 11, 21-23]。在基础实验动物模型中,Trastuzumab可明显抑制HER2阴性的 Luminal 型乳腺癌细胞在小鼠上的肿瘤增殖生长,这暗示了Trastuzumab可用于HER2阴性乳腺癌治疗的潜在可能性[24]。对NSABPB-31 and NCCTGN9831两项临床试验回顾性分析发现:HER2低表达乳腺癌患者可从Trastuzumab治疗中生存获益[25]。然而,NSABPB-47临床试验发现HER2低表达乳腺癌患者,并未从Trastuzumab靶向治疗中获益[26]。以上研究揭示了Trastuzumab对HER2低表达乳腺癌患者临床效果并不显著。帕妥珠单抗(Pertuzumab)通过与HER2蛋白细胞外侧段的 II 区结合,发挥其功能作用。但是,仅使用Pertuzumab治疗HER2低表达乳腺癌的临床试验,并未达到预期试验终点[27]

(2)抗体偶联药物(Antibody-Drug Conjugates,ADC)

近年来,“魔法子弹”ADC药物在乳腺癌治疗中技惊四座,为乳腺癌患者的治疗带来新曙光。其中,曲妥珠单抗-美坦新偶联物(Ado-trastuzumab emtansine,T-DM1)是由Trastuzumab和细胞毒性药物美坦新通过连接子偶联而成的新型ADC药物,是最早研发的抗HER2 ADC药物,也是我国首个上市的治疗乳腺癌ADC药物,可特异性地将细胞毒性药物靶向释放入HER2阳性乳腺癌细胞内,可针对性地杀死乳腺癌细胞,发挥高效抗肿瘤作用。2013年2月22日,美国FDA批准T-DM1用于既往接受过Trastuzumab和紫杉烷单独或联合治疗的HER2阳性转移性乳腺癌。2019年5月3日,美国FDA批准T-DM1用于接受紫杉烷和Trastuzumab新辅助治疗后有残留病灶的HER2阳性早期乳腺癌患者的辅助强化治疗[28]。但是,针对HER2 低表达乳腺癌患者,两项单臂的II期临床研究发现:

T-DM1对HER2 低表达乳腺癌患者的临床治疗效果不尽人意,这可能是因为T-DM1对旁边的乳腺癌细胞膜穿透能力较差所致[14, 29]
目前,新型ADC药物的曲妥珠单抗-德鲁替康(Trastuzumab deruxtecan,T-DXd,DS-8201),不仅在HER2阳性乳腺癌中表现出喜人的抗肿瘤效果,也适用于HER2低表达乳腺癌[30]。T-DXd是由HER2抗体、拓扑异构酶I抑制剂通过可裂解的特异酶切连接子偶联而成。T-DXd不仅可以抑制HER2蛋白,还可通过其高细胞膜穿透性渗透至周围的乳腺癌细胞内,产生旁观者效应。

图片14.png图3. T-DXd---新一代ADC药物设计及特点

根据III期DESTINY-Breast 04临床试验,T-DXd在曾接受治疗的HER2低表达晚期乳腺癌中的ORR超过50%[31]。针对既往经历过多线治疗失败的晚期 HER2低表达乳腺癌患者,T-DXd仍然疗效斐然[30, 32, 33]。因此,2023年7月12日,国家药品监督管理局(NMPA)批准T-DXd单药用于治疗既往在转移性疾病阶段接受过至少一种系统治疗的,或完成辅助化疗之后6个月内复发的,不可切除或转移性HER2低表达乳腺癌的适应症获批。2023年ESMO晚期乳腺癌指南推荐,CDK4/6抑制剂联合内分泌一线治疗针对快速进展及靶向联合内分泌两线经治人群就,都推荐优选T-DXd[34]。2023年V4版NCCN指南指出:内脏危象或内分泌难治的HR+/HER2低表达晚期乳腺癌患者,二线优选T-DXd(1类证据);T-DXd同时也是晚期三阴性乳腺癌(TNBC)二线治疗的1类优选推荐方案[35]

图片15.png图4. 2023年ESMO指南关于HR+/HER2低表达晚期乳腺癌的治疗[34]

图片16.png图5. 2023年V4版NCCN指南关于HER2低表达乳腺癌的治疗推荐[35]

曲妥珠单抗-杜卡霉素(Trastuzumab duocarmazin,SYD985)也是靶向HER2的新型ADC药物,将Trastuzumab与杜卡霉素(vc-seco-DUBA)进行偶联。PDX模型抗肿瘤活性研究揭示了 SYD985在HER2低表达中也显著疗效[36]。同时,SYD985治疗HER2低表达晚期乳腺癌I期临床试验表明:乳腺癌患者中位PFS为9.4个月,ORR可达到33%[9]。维迪西妥单抗(disitamab vedotin,RC48)是第一个国产新型HER2靶点的ADC药物,将赫妥珠单抗(hertuzumab)与单甲基奥瑞他汀 E(monomethyl auristatin E,MMAE)偶联[37]。RC48在HER-2低表达PDX模型的抗肿瘤作用明显强于Trastuzumab。RC48在HER2低表达晚期乳腺癌患者的临床试验研究显示:HER2低表达乳腺癌患者的中位PFS为5.7个月,ORR为39.6%[38]。目前,RC48-ADC的临床试验正在紧锣密鼓开展中[39, 40]

(3)其他药物及临床试验

除了上述药物,目前全世界的科学家门,也在积极研发其他新型ADC药物(ARX788、A166等)、双特异性抗体(KN026、ZW25、ertumaxomab等)和乳腺癌疫苗(nelipepimut-S、GP2、AE37等)[41, 42]

4.总结

既往多项研究在HER2低表达乳腺癌中折戟沉沙,但T-DXd凭借其强大的治疗效果,显著改善HER2低表达乳腺癌的生存预后,扭转了HER2低表达乳腺癌靶向治疗探索的颓势。尽管,目前尚缺乏充足循证医学证据,将HER2低表达乳腺癌归为一个独立的新亚型。我科真实世界研究,对HER2低表达乳腺癌提供新的治疗策略方向。我们的研究发现HER2低表达乳腺癌具有PI3K-AKT通路相关基因的多个重要突变,那么是否可在新辅助治疗中予化疗联合PI3K-AKT通路的靶向治疗,以提高其pCR率,也是值得进一步探索的方向。我们也期待未来有一系列的高质量研究,将为HER2低表达乳腺癌治疗领域夯实基础,为HER2低表达乳腺癌患者带来更大生存获益,使HER2低表达乳腺癌迎来里程碑式的精准治疗时代。


参考文献

1. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023; 73: 17-48.
2. Li J, Zhou J, Wang H, Liu Z, Fan Z, Liu Y, et al. Trends in Disparities and Transitions of Treatment in Patients With Early Breast Cancer in China and the US, 2011 to 2021. JAMA Netw Open. 2023; 6: e2321388.
3. Waks AG, Winer EP. Breast Cancer Treatment: A Review. JAMA. 2019; 321: 288-300.
4. Yap Y-S, Lu Y-S, Tamura K, Lee JE, Ko EY, Park YH, et al. Insights Into Breast Cancer in the East vs the West. JAMA Oncology. 2019; 5: 1489-96.
5. Cao W, Chen HD, Yu YW, Li N, Chen WQ. Changing profiles of cancer burden worldwide and in China: a secondary analysis of the global cancer statistics 2020. Chin Med J (Engl). 2021; 134: 783-91.
6. Gampenrieder SP, Rinnerthaler G, Tinchon C, Petzer A, Balic M, Heibl S, et al. Landscape of HER2-low metastatic breast cancer (MBC): results from the Austrian AGMT_MBC-Registry. Breast Cancer Res. 2021; 23: 112.
7. Tarantino P, Hamilton E, Tolaney SM, Cortes J, Morganti S, Ferraro E, et al. HER2-Low Breast Cancer: Pathological and Clinical Landscape. J Clin Oncol. 2020; 38: 1951-62.
8. Denkert C, Seither F, Schneeweiss A, Link T, Blohmer J-U, Just M, et al. Clinical and molecular characteristics of HER2-low-positive breast cancer: pooled analysis of individual patient data from four prospective, neoadjuvant clinical trials. The Lancet Oncology. 2021; 22: 1151-61.
9. Banerji U, van Herpen CML, Saura C, Thistlethwaite F, Lord S, Moreno V, et al. Trastuzumab duocarmazine in locally advanced and metastatic solid tumours and HER2-expressing breast cancer: a phase 1 dose-escalation and dose-expansion study. Lancet Oncol. 2019; 20: 1124-35.
10. Swain SM, Baselga J, Kim SB, Ro J, Semiglazov V, Campone M, et al. Pertuzumab, trastuzumab, and docetaxel in HER2-positive metastatic breast cancer. N Engl J Med. 2015; 372: 724-34.
11. Pivot X, Romieu G, Debled M, Pierga JY, Kerbrat P, Bachelot T, et al. 6 months versus 12 months of adjuvant trastuzumab in early breast cancer (PHARE): final analysis of a multicentre, open-label, phase 3 randomised trial. Lancet. 2019; 393: 2591-8.
12. Hurvitz SA, Martin M, Jung KH, Huang CS, Harbeck N, Valero V, et al. Neoadjuvant Trastuzumab Emtansine and Pertuzumab in Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer: Three-Year Outcomes From the Phase III KRISTINE Study. J Clin Oncol. 2019; 37: 2206-16.
13. Swain SM, Baselga J, Kim SB, Ro J, Semiglazov V, Campone M, et al. Pertuzumab, trastuzumab, and docetaxel in HER2-positive metastatic breast cancer. N Engl J Med. 2015; 372: 724-34.
14. Krop IE, LoRusso P, Miller KD, Modi S, Yardley D, Rodriguez G, 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 Clin Oncol. 2012; 30: 3234-41.
15. Wolff AC, Hammond MEH, Allison KH, Harvey BE, Mangu PB, Bartlett JMS, et al. Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Focused Update. J Clin Oncol. 2018; 36: 2105-22.
16. Tarantino P, Viale G, Press MF, Hu X, Penault-Llorca F, Bardia A, et al. ESMO expert consensus statements (ECS) on the definition, diagnosis, and management of HER2-low breast cancer. Ann Oncol. 2023; 34: 645-59.
17. Schettini F, Chic N, Brasó-Maristany F, Paré L, Pascual T, Conte B, et al. Clinical, pathological, and PAM50 gene expression features of HER2-low breast cancer. npj Breast Cancer. 2021; 7: 1.
18. Horisawa N, Adachi Y, Takatsuka D, Nozawa K, Endo Y, Ozaki Y, et al. The frequency of low HER2 expression in breast cancer and a comparison of prognosis between patients with HER2-low and HER2-negative breast cancer by HR status. Breast Cancer. 2022; 29: 234-41.
19. Peiffer DS, Zhao F, Chen N, Hahn OM, Nanda R, Olopade OI, et al. Clinicopathologic Characteristics and Prognosis of ERBB2-Low Breast Cancer Among Patients in the National Cancer Database. JAMA Oncology. 2023; 9: 500-10.
20. Zhang G, Ren C, Li C, Wang Y, Chen B, Wen L, et al. Distinct clinical and somatic mutational features of breast tumors with high-, low-, or non-expressing human epidermal growth factor receptor 2 status. BMC Med. 2022; 20: 142.
21. Dang C, Iyengar N, Datko F, D'Andrea G, Theodoulou M, Dickler M, et al. Phase II study of paclitaxel given once per week along with trastuzumab and pertuzumab in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer. J Clin Oncol. 2015; 33: 442-7.
22. Untch M, Fasching PA, Konecny GE, Hasmuller S, Lebeau A, Kreienberg R, et al. Pathologic complete response after neoadjuvant chemotherapy plus trastuzumab predicts favorable survival in human epidermal growth factor receptor 2-overexpressing breast cancer: results from the TECHNO trial of the AGO and GBG study groups. J Clin Oncol. 2011; 29: 3351-7.
23. Jeon SH, Shin KH, Kim JH, Kim K, Kim IA, Lee KH, et al. Effects of trastuzumab on locoregional recurrence in human epidermal growth factor receptor 2-overexpressing breast cancer patients treated with chemotherapy and radiotherapy. Breast Cancer Res Treat. 2018; 172: 619-26.
24. Ithimakin S, Day KC, Malik F, Zen Q, Dawsey SJ, Bersano-Begey TF, et al. HER2 drives luminal breast cancer stem cells in the absence of HER2 amplification: implications for efficacy of adjuvant trastuzumab. Cancer Res. 2013; 73: 1635-46.
25. Perez EA, Romond EH, Suman VJ, Jeong JH, Sledge G, Geyer CE, Jr., et al. Trastuzumab plus adjuvant chemotherapy for human epidermal growth factor receptor 2-positive breast cancer: planned joint analysis of overall survival from NSABP B-31 and NCCTG N9831. J Clin Oncol. 2014; 32: 3744-52.
26. Fehrenbacher L, Cecchini RS, Geyer CE, Jr., Rastogi P, Costantino JP, Atkins JN, et al. NSABP B-47/NRG Oncology Phase III Randomized Trial Comparing Adjuvant Chemotherapy With or Without Trastuzumab in High-Risk Invasive Breast Cancer Negative for HER2 by FISH and With IHC 1+ or 2. J Clin Oncol. 2020; 38: 444-53.
27. Gianni L, Lladó A, Bianchi G, Cortes J, Kellokumpu-Lehtinen PL, Cameron DA, et al. Open-label, phase II, multicenter, randomized study of the efficacy and safety of two dose levels of Pertuzumab, a human epidermal growth factor receptor 2 dimerization inhibitor, in patients with human epidermal growth factor receptor 2-negative metastatic breast cancer. J Clin Oncol. 2010; 28: 1131-7.
28. von Minckwitz G, Huang CS, Mano MS, Loibl S, Mamounas EP, Untch M, et al. Trastuzumab Emtansine for Residual Invasive HER2-Positive Breast Cancer. N Engl J Med. 2019; 380: 617-28.
29. Burris HA, 3rd, Rugo HS, Vukelja SJ, Vogel CL, Borson RA, Limentani S, et al. Phase II study of the antibody drug conjugate trastuzumab-DM1 for the treatment of human epidermal growth factor receptor 2 (HER2)-positive breast cancer after prior HER2-directed therapy. J Clin Oncol. 2011; 29: 398-405.
30. Modi S, Saura C, Yamashita T, Park YH, Kim SB, Tamura K, et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Breast Cancer. N Engl J Med. 2020; 382: 610-21.
31. Modi S, Jacot W, Yamashita T, Sohn J, Vidal M, Tokunaga E, et al. Trastuzumab Deruxtecan in Previously Treated HER2-Low Advanced Breast Cancer. New England Journal of Medicine. 2022; 387: 9-20.
32. Doi T, Shitara K, Naito Y, Shimomura A, Fujiwara Y, Yonemori K, 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. Lancet Oncol. 2017; 18: 1512-22.
33. Modi S, Park H, Murthy RK, Iwata H, Tamura K, Tsurutani J, 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 Clin Oncol. 2020; 38: 1887-96.
34. Curigliano G, Castelo-Branco L, Gennari A. ESMO Metastatic Breast Cancer Living Guidelines, v1.1 May 2023.
35. NCCN clinical practice guidelines in oncology(NCCN Guidelines®); Breast Cancer, Version 42023—March 23,2023.
36. van der Lee MM, Groothuis PG, Ubink R, van der Vleuten MA, van Achterberg TA, Loosveld EM, et al. The Preclinical Profile of the Duocarmycin-Based HER2-Targeting ADC SYD985 Predicts for Clinical Benefit in Low HER2-Expressing Breast Cancers. Mol Cancer Ther. 2015; 14: 692-703.
37. Li L, Xu MZ, Wang L, Jiang J, Dong LH, Chen F, et al. Conjugating MMAE to a novel anti-HER2 antibody for selective targeted delivery. Eur Rev Med Pharmacol Sci. 2020; 24: 12929-37.
38. Wang J, Liu Y, Zhang Q, al e. RC48-ADC,a HER2-targetingantibody-drug conjugate, in patients with HER2-positive and HER2-lowexpressing advanced or metastatic breast cancer: A pooled analysis of two studies. J Clin Oncol,2021,39( 15_suppl) : 1022. 2021.
39. Xu Y, Wang Y, Gong J, Zhang X, Peng Z, Sheng X, et al. Phase I study of the recombinant humanized anti-HER2 monoclonal antibody-MMAE conjugate RC48-ADC in patients with HER2-positive advanced solid tumors. Gastric Cancer. 2021; 24: 913-25.
40. Shi F, Liu Y, Zhou X, Shen P, Xue R, Zhang M. Disitamab vedotin: a novel antibody-drug conjugates for cancer therapy. Drug Deliv. 2022; 29: 1335-44.
41. Mittendorf EA, Lu B, Melisko M, Price Hiller J, Bondarenko I. Efficacy and Safety Analysis of Nelipepimut-S Vaccine to Prevent Breast Cancer Recurrence: A Randomized, Multicenter, Phase III Clinical Trial. Clin Cancer Res. 2019; 25: 4248-54.
42. Mittendorf EA, Ardavanis A, Symanowski J, Murray JL, Shumway NM, Litton JK, et al. Primary analysis of a prospective, randomized, single-blinded phase II trial evaluating the HER2 peptide AE37 vaccine in breast cancer patients to prevent recurrence. Ann Oncol. 2016; 27: 1241-8.