您好,欢迎您

【35under35】韦青医生:ADC药物旁观者效应在胃癌治疗中的关键意义

2022年07月30日
作者:韦青
医院:中国科学院大学附属肿瘤医院

 

               
韦青
主治医师,医学博士

中国科学院大学附属肿瘤医院(浙江省肿瘤医院) 肝胆胰胃内科 主治医师 医学博士(博士后在站)
CSCO青年专家委员会委员
CSCO青年专家委员会秘书
CSCO青委翻译小组成员
浙江省抗癌协会肿瘤内科青委会委员
浙江省肿瘤精准治疗专委会青委会秘书
《肿瘤综合治疗杂志电子版》编委
JCO中文版-消化肿瘤专刊编委会青年编委
浙江省肿瘤医院人才创新“30”培养对象
于北京大学医学部获得博士研究生学位,研究生阶段师从王洁教授,博士阶段师从沈琳教授
主要从事胃肠道肿瘤的综合诊疗及临床转化研究,一作/通讯作者发表SCI论文9篇,其中10分以上2篇,主持及参与省级以上课题多项

一、晚期胃癌抗HER2治疗面临巨大未满足的临床需求

胃癌在我国发病率居第2位,严重危害我国人民健康1。与此同时,60%-70%患者就诊时已为中晚期,丧失了最佳手术机会,由于缺乏有效的靶向药物,这类患者预后较差。晚期胃癌患者中有17%~20%的比例存在人类表皮生长因子2(Human epidermal growth factor 2,HER2)过表达或者扩增2。2010年全球、多中心、III期ToGA研究(中位OS为13.8个月和11.1个月)证实了化疗+曲妥珠单抗(Trastuzumab)是HER2过表达(阳性)胃癌的标准一线治疗方案,延长了患者的生存期3。此后10年,国际上对于HER2过表达胃癌的探索从未止步,然而追寻靶向药物之路异常曲折,2015年的LOGiC研究(Lapatinib)、2017年的Gatsby研究(T-DM1)、2018年的JACOB研究(Trastuzumab + Pertuzumab)等探索HER2靶点新药的临床研究均折戟沉沙4,5,6,尤其是乳腺癌中疗效甚好的抗体偶联药物(antibody-drug conjugate,ADC)恩美曲妥珠单抗(ado-trastuzumab emtansine, T-DM1)在胃癌中却疗效甚微5,7,患者生存获益非常有限。

胃癌抗HER2治疗不同于乳腺癌的一个显著特征就是具有更加异质化的HER2表达8,9,即便是HER2高表达的胃癌,其肿瘤内部和周围仍会存在大量HER2低表达的胃癌细胞及其他细胞。如此高度异质化的肿瘤靶点表达显著增加了胃癌靶向治疗的难度。因此,晚期胃癌抗HER2治疗仍然面临巨大且未满足的临床需求。

二、新一代ADC药物因为“旁观者效应”在HER2强异质性的胃癌中发挥出有效杀伤作用

抗体偶联药物(antibody-drug conjugate,ADC)由单克隆抗体、连接子和小分子细胞毒素偶联而成。从机制上来说,ADC药物进入体内后,抗体部分与表达抗原的靶细胞特异性结合后介导ADC内吞,进入溶酶体进行降解后,小分子细胞毒弹头以游离形式被释放进入细胞质,通过抑制细胞微管形成或DNA拓扑异构酶,完成对癌细胞的杀伤10。ADC药物的优点是结合了抗体药物的高特异靶向性和小分子细胞毒素的高细胞杀伤力,实现对肿瘤的精准化疗,目前已经成为抗肿瘤药物研发的重点方向。然而,靶向HER2的ADC药物T-DM1早在2017年Gatsby研究已报道在HER2阳性(HER2+)胃癌二线治疗中无明显疗效5。直到2020年的Destiny-GC01研究公布了Trastuzumab deruxtecan (T-DXd, DS-8201) 在胃癌中用于至少经两种治疗进展的HER2过表达晚期患者的疗效和安全性,研究结果发表在新英格兰医学期刊11。与其他三线治疗或后线治疗方案相比,T-DXd明显提高了晚期患者的ORR(51% vs 14%),延长了患者的OS(12.5月 vs 8.4月,P=0.01)11。2021年,我国原创的HER2靶向ADC药物维迪西妥单抗(Disitamab vedotin, RC48)也在HER2过表达晚期肿瘤中展现出有效性和安全性,并获批用于临床治疗12,13

虽然T-DXd在针对HER2过表达或HER2突变实体瘤临床治疗中取得突出的疗效,背后起到关键作用的ADC药物“旁观者效应”的机制尚未完全阐明。T-DXd与T-DM1相比,是新一代的抗体偶联药物,由重组人源化抗HER2 IgG1单克隆抗体Trastuzumab、酶切裂解型GGFG肽基连接子和拓扑异构酶Ⅰ抑制剂DXd弹头组成14(图2)。虽然同为HER2靶向ADC药物,T-DXd的临床治疗效果显著优于T-DM1,最相关的特征是其“旁观者效应”15。HER2+细胞介导T-DXd内化后,其酶切型可裂解连接子(GGFG四肽)被肿瘤细胞内体和溶酶体中高表达的组织蛋白酶Cathepsin B裂解,释放出大量DNA拓扑异构酶抑制剂DXd弹头。在杀死靶细胞后,游离的DXd弹头会从靶细胞渗出继续杀伤肿瘤微环境内相邻的HER2阴性(HER2-)细胞,即旁观者效应16。上一代ADC药物(如T-DM1)由于采用不可裂解型SMCC连接子,因此不具备旁观者效应,对于HER2异质性强的胃癌难以获得有效的临床疗效5。由此可见,“旁观者效应”是ADC药物发挥疗效的重要基础,厘清其具体发生机制是一个关键科学问题。

三、旁观者效应是药物独特设计和作用机制的综合结果

 影响旁观者效应的因素有很多。前文提到的T-DM1和目前在诸多瘤种中大获全胜的T-DXd是最典型的例子。T-DXd(又名DS-8201a)由曲妥珠单抗(靶向HER2)、毒素DXd(拓扑异构酶I抑制剂)、四肽连接子构成,其中四肽连接子与抗体的半胱氨酸残基进行连接(图2)。T-DM1由曲妥珠单抗、毒素DM1(微管蛋白聚合抑制剂)、硫醚连接子(不可裂解)构成(图2)。抗体与连接子之间化学键的稳定性是影响关键因素之一,连接子通常分为可裂解和不可裂解,前者包括化学不稳定连接(如对pH敏感的腙键、二硫键)和酶不稳定连接(如二肽,可被组蛋白酶B切割),后者主要为共价连接(如硫醚键),必须内化进入细胞,由溶酶体对其进行分解,才能释放毒素。

细胞毒素的性质也是影响旁观者效应的重要因素,极性小的毒素容易透过细胞膜,因此在肿瘤细胞中释放后仍有可能通过胞膜扩散到肿瘤外,从而作用于周围的细胞,产生旁观者效应,而极性太大的毒素则难以穿透细胞膜。这里也有特殊情况,例如T-DM1虽然其细胞毒素极性并没有很大,但由于其裂解不完全,导致毒素上残留一个带正电的赖氨酸,这使其无法透过细胞膜,因此T-DM1不会产生旁观者效应。DXd具有较好的透膜性,产生旁观者效应。此外,当目标肿瘤细胞最终瓦解时,其内部的细胞毒素会被释放出来,扩散到周围的细胞,发挥旁观者效应(图1)。但需要注意的是:药物作为一个整体,进入到复杂的肿瘤内部,不能孤立地看待其设计及作用机制,旁观者效应是药物与环境各种机制的综合结果。

1.png

doi: 10.1038/bjc.2017.367(图1)

屏幕截图 2022-07-30 115802.png

不可裂解型T-DM1与酶切裂解型T-DXd的结构对比(图2由选手绘制)

四、ADC药物的特殊毒性如间质性肺炎是否与旁观者效应相关?

对于大多数靶向HER2的药物,已经观察到一定程度的肺毒性,尽管频率和严重程度各不相同。在最近的一项包括近10,000名抗HER2药物治疗的晚期乳腺癌系统评价中,肺毒性总发生率为2.4%,其中0.2%(n = 16)导致死亡17。若局限在抗HER2 ADC,T-DM1的 ILD的发生率相对较低,3290名患者中只有15名(0.5%)患有ILD,6名患者(0.2%)发生了ILD相关的死亡。

在对542名接受T-Dxd治疗的癌症患者(80%患有晚期乳腺癌)的汇总分析中,16.8%经历过任一级别的ILD,中位发病时间为208天(范围:0-582天),并确定了一些潜在的危险因素,包括更高的药物剂量、日本血统, ILD 的发生率在 6.4 mg/kg (19.5%) 时更高18,19

抗体偶联药物相关的肺损伤理论上可能与(1)ADC的靶点依赖性摄取有关,(2)正常细胞中ADC的靶点依赖性摄取,(3)癌细胞释放的游离弹头杀伤旁观者细胞,以及(4)ADC未能偶联弹头产生的循环自由弹头(图3)。一些实验观察表明,不依赖靶点的摄取可能部分解释肺部损伤。最近的一项研究在食蟹猴中以不同剂量注射3个月T-Dxd或非结合形式的有效载荷 (deruxtecan)。在接受T-Dxd的食蟹猴中,以剂量依赖性地形式观察到了典型的组织病理学特征的ILD的发作,而注射deruxtecan即使剂量升高也不会导致肺毒性。而食蟹猴HER2的表达类似于人类仅限于支气管上皮。然而,肺损伤主要在肺泡水平上发现,T-Dxd也是被发现局限于肺泡巨噬细胞而不是在肺上皮细胞中20

4.png

doi: 10.1001/jamaoncol.2021.3595.(图3)

虽然还未有进一步体内验证以及临床数据,但这些发现支持了这样一种假设,即循环中未结合的弹头可能无法解释T-Dxd观察到的肺毒性作用。且新一代ADC药物的肺毒性显著强于第二代ADC药物(表1),ADC诱导的肺泡损伤可能与免疫细胞对ADC的靶点非依赖性摄取有关,这可能也是旁观者效应在肺损伤中的作用机制之一。可并非所有的新型ADC药物都有该现象。虽然不排除样本量的原因,RC48作为国产新药并没有观察到ILD的发生。

表1.部分抗HER2 靶点ADC的药物ILD发生率屏幕截图 2022-07-30 120153.png

五、总结

随着目前 ADC 药物合成技术的突破,逐步成为新药研发的热门方向,多款 ADC 药物被 FDA 批准上市,不同靶点、连接子、弹头的设计不仅仅需要考虑疗效,还要考虑安全性。对 ADC 药物背后机制理解的重要性和迫切性也逐渐凸显。期待未来有更多的探索,能够让 ADC 药物更好地服务于临床。

参考文献

[1] Wanqing Chen, Rongshou Zheng, Peter D Baade, Siwei Zhang, Hongmei Zeng, Freddie Bray, Ahmedin Jemal, Xue Qin Yu, Jie He. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66(2):115-32.

[2] Elizabeth C Smyth, Magnus Nilsson, Heike I Grabsch, Nicole Ct van Grieken, Florian Lordick. Gastric cancer. Lancet. 2020;396(10251):635-48.

[3] Yung-Jue Bang, Eric Van Cutsem, Andrea Feyereislova, Hyun C Chung, Lin Shen, Akira Sawaki, Florian Lordick, Atsushi Ohtsu, Yasushi Omuro, Taroh Satoh, Giuseppe Aprile, Evgeny Kulikov, Julie Hill, Michaela Lehle, Josef Rüschoff, Yoon-Koo Kang, ToGA Trial Investigators. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 2010;376(9742):687-97.

[4] J Randolph Hecht, Yung-Jue Bang, Shukui K Qin, Hyun C Chung, Jianming M Xu, Joon O Park, Krzysztof Jeziorski, Yaroslav Shparyk, Paulo M Hoff, Alberto Sobrero, Pamela Salman, Jin Li, Svetlana A Protsenko, Zev A Wainberg, Marc Buyse, Karen Afenjar, Vincent Houé, Agathe Garcia, Tomomi Kaneko, Yingjie Huang, Saba Khan-Wasti, Sergio Santillana, Michael F Press, Dennis Slamon. Lapatinib in Combination With Capecitabine Plus Oxaliplatin in Human Epidermal Growth Factor Receptor 2-Positive Advanced or Metastatic Gastric, Esophageal, or Gastroesophageal Adenocarcinoma: TRIO-013/LOGiC--A Randomized Phase III Trial. J Clin Oncol. 2016;34(5):443-51.

[5] Peter C Thuss-Patience, Manish A Shah, Atsushi Ohtsu, Eric Van Cutsem, Jaffer A Ajani, Hugo Castro, Wasat Mansoor, Hyun Cheol Chung, Gyorgy Bodoky, Kohei Shitara, Gail D Lewis Phillips, Tina van der Horst, Marie-Laurence Harle-Yge, Betsy L Althaus, Yoon-Koo Kang.  Trastuzumab emtansine versus taxane use for previously treated HER2-positive locally advanced or metastatic gastric or gastro-oesophageal junction adenocarcinoma (GATSBY): an international randomised, open-label, adaptive, phase 2/3 study. Lancet Oncol. 2017;18(5):640-53.

[6] Josep Tabernero, Paulo M Hoff, Lin Shen, Atsushi Ohtsu, Manish A Shah, Karen Cheng, Chunyan Song, Haiyan Wu, Jennifer Eng-Wong, Katherine Kim, Yoon-Koo Kang. Pertuzumab plus trastuzumab and chemotherapy for HER2-positive metastatic gastric or gastro-oesophageal junction cancer (JACOB): final analysis of a double-blind, randomised, placebo-controlled phase 3 study. Lancet Oncol. 2018;19(10):1372-84.

[7] Sunil Verma, David Miles, Luca Gianni, Ian E Krop, Manfred Welslau, José Baselga, Mark Pegram, Do-Youn Oh, Véronique Diéras, Ellie Guardino, Liang Fang, Michael W Lu, Steven Olsen, Kim Blackwell, EMILIA Study Group. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med. 2012;367(19):1783-91.

[8] Josef Rüschoff, Wedad Hanna, Michael Bilous, Manfred Hofmann, Robert Y Osamura, Frédérique Penault-Llorca, Marc van de Vijver, Giuseppe Viale. HER2 testing in gastric cancer: a practical approach. Mod Pathol. 2012;25(5):637-50.

[9] VS Warneke, H-M Behrens, C Böger, T Becker, F Lordick, M P A Ebert, C Röcken. Her2/neu testing in gastric cancer: evaluating the risk of sampling errors. Ann Oncol. 2013;24(3):725-33.

[10] Joshua Z Drago, Shanu Modi, Sarat Chandarlapaty. Unlocking the potential of antibody-drug conjugates for cancer therapy. Nat Rev Clin Oncol. 2021;18(6):327-44.

[11] Kohei Shitara, Yung-Jue Bang, Satoru Iwasa, Naotoshi Sugimoto, Min-Hee Ryu, Daisuke Sakai, Hyun-Cheol Chung, Hisato Kawakami, Hiroshi Yabusaki, Jeeyun Lee, Kaku Saito, Yoshinori Kawaguchi, Takahiro Kamio, Akihito Kojima, Masahiro Sugihara, Kensei Yamaguchi, DESTINY-Gastric01 Investigators. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Gastric Cancer. N Engl J Med. 2020;382(25):2419-30.

[12] Yingying Xu, Yakun Wang , Jifang Gong, Xiaotian Zhang, Zhi Peng, Xinan Sheng, Chenyu Mao, Qingxia Fan, Yuxian Bai, Yi Ba, Da Jiang, Fen Yang, Changsong Qi, Jian Li, Xicheng Wang, Jun Zhou, Ming Lu, Yanshuo Cao, Jiajia Yuan, Dan Liu, Zhenghang Wang, Jianmin Fang, Lin Shen. 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(4):913-25.

[13] Xinan Sheng, Xieqiao Yan, Lin Wang, Yanxia Shi, Xin Yao, Hong Luo, Benkang Shi, Jiyan Liu, Zhisong He, Guohua Yu, Jianming Ying, Weiqing Han, Changlu Hu, Yun Ling, Zhihong Chi, Chuanliang Cui, Lu Si, Jianmin Fang, Aiping Zhou, Jun Guo. Open-label, Multicenter, Phase II Study of RC48-ADC, a HER2-Targeting Antibody-Drug Conjugate, in Patients with Locally Advanced or Metastatic Urothelial Carcinoma. Clin Cancer Res. 2021;27(1):43-51.

[14] Ogitani Y, Aida T, Hagihara K et al. DS-8201a, a novel HER2-targeting ADC with a novel DNA topoisomerase I inhibitor, demonstrates a promising anti-tumor efficacy with differentiation from T-DM1. Clin Cancer Res. 2016;22(20):5097-108.

[15] Ogitani Y, Hagihara K, Oitate M, Naito H, Agatsuma T. Bystander killing effect of DS-8201a, a novel anti-human epidermal growth factor receptor 2 antibody-drug conjugate, in tumors with human epidermal growth factor receptor 2 heterogeneity. Cancer Sci. 2016;107:1039–46.

[16] Li F, Emmerton KK, Jonas M, Zhang X, Miyamoto JB, Setter JR, Nicholas ND,Okeley NM, Lyon RP, Benjamin DR, Law CL (2016) intracellular released payload influences potency and bystander-killing effects of antibody-drug conjugates in preclinical models. Cancer Res.76(9): 2710–19.

[17] HackshawMD, Danysh HE, Singh J, et al. Incidence of pneumonitis/interstitial lung disease induced by HER2-targeting therapy for HER2-positivemetastatic breast cancer. Breast Cancer Res Treat. 2020;183(1):23-39.

[18] Powell C, Camidge D, Gemma A, et al.Characterization, monitoring and management of interstitial lung disease in patients with metastatic breast cancer: analysis of data available from multiple studies of DS-8201a, a HER2-targeted ADC

with a topoisomerase I inhibitor payload. San Antonio Breast Cancer Symposium; December 4-8, 2018.

[19] Powell CA, Camidge DR, Modi S, et al. Risk factors for interstitial lung disease in patients treated with trastuzumab deruxtecan from two interventional studies [abstract]. Ann Oncol. 2020; 31(suppl 4):289P.

[20] Kumagai K, Aida T, Tsuchiya Y, Kishino Y, Kai K, Mori K. Interstitial pneumonitis related to trastuzumab deruxtecan, a human epidermal growth factor receptor 2-targeting Ab-drug conjugate, in monkeys. Cancer Sci. 2020;111(12):

4636-4645.

[21] von Minckwitz G, Huang CS, Mano MS, et al.Trastuzumab emtansine for residual invasive HER2-positive breast cancer. N Engl J Med. 2019; 380(7):617-628. 2018;1-12.

[22] Emens LA, Esteva FJ, Beresford M, et al. Trastuzumab emtansine plus atezolizumab versus trastuzumab emtansine plus placebo in previously treated, HER2-positive advanced breast cancer (KATE2): a phase 2, multicentre, randomised,

double-blind trial. Lancet Oncol.

[23] Modi S, Saura C, Yamashita T, et al. Updated results from DESTINY-Breast01, a phase 2 trial of trastuzumab deruxtecan (T-DXd) in HER2 positive metastatic breast cancer [abstract]. Cancer Res.

[24] Shitara K, Bang Y-J, Iwasa S, et al; DESTINY-Gastric01 Investigators. Trastuzumab deruxtecan in previously treated HER2-positive gastric cancer. N Engl J Med. 2020;382(25):2419-2430.

[25] Li BT, Smit EF, Goto Y, et al. Trastuzumab deruxtecan in HER2-mutant non–small-cell lung cancer. N Engl J Med. Published online September 18, 2021.

[26] Siena S, Di Bartolomeo M, Raghav KPS, et al.A phase II, multicenter, open-label study of trastuzumab deruxtecan (T-DXd; DS-8201) in patients (pts) with HER2-expressingmetastatic colorectal cancer (mCRC): DESTINY-CRC01[abstract]. J Clin Oncol. 2020;38(15)(suppl):4000.

[27] Cortes J, Kim S-B, ChungW-P, et al.LBA1—trastuzumab deruxtecan (T-DXd) vs

trastuzumab emtansine (T-DM1) in patients (Pts) with HER2+ metastatic breast cancer (mBC): results of the randomized phase III DESTINY-Breast03 study. Ann Oncol. 2021;32(suppl 5):S1283-S1346.

[28] Hamilton EP, Shapiro C, Petrylak D, et al.Trastuzumab deruxtecan (T-DXd; DS-8201) with nivolumab in patients with HER2-expressing, advanced breast cancer: a 2-part, phase 1b,multicenter, open-label study. Cancer Res. 2021;81(suppl 4):PD3-PD07.

[29] Banerji U, van Herpen CML, Saura C, 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(8): 1124-1135.

[30] Saura C, O’Shaughnessy J, Aftimos P, et al.LBA15—primary outcome of the phase III SYD985.002/TULIP trial comparing [vic-]trastuzumab duocarmazine to physician’s choice treatment in patients with pre-treated HER2-positive locally advanced or metastatic breast cancer. Ann Oncol. 2021;32(suppl 5): S1283-S1346.

[31] Yingying Xu, Yakun Wang, Jifang Gong , 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 Jul;24(4):913-925.