Research progress on the use of an antibody–drug conjugate, DS-8201a, in advanced breast cancer
-
摘要: 抗体药物偶联物(antibody-drug conjugates,ADCs)是一种由抗体、细胞毒药物和偶联链组成的新型抗肿瘤制剂,具有良好的靶向性及抗癌活性。对于既往经过多线治疗失败的人表皮生长因子受体-2(human epidermal growth factor receptor-2,HER-2)阳性晚期乳腺癌,新一代ADCs制剂DS-8201a在疾病控制率、改善生存等方面具有良好的治疗效果,并对HER-2低表达的晚期乳腺癌患者亦带来生存获益,而药物相关的血液学毒性、间质性肺疾病等安全性问题也引起了广泛关注。本文将就DS-8201a在晚期乳腺癌中的研究进展进行综述。Abstract: Antibody–drug conjugates (ADCs) represent a new class of antitumor agents composed of an antibody, a cytotoxic drug, and a flexible linker, which together provide highly tumor-specific targeting and potent antitumor activity. For patients with human epidermal growth factor receptor-2 (HER-2)-positive advanced breast cancer who have failed to respond to previous multiline therapies, the next-generation ADC, DS-8201a, delivers a good therapeutic effect as measured by disease control rate and improved survival. It also provides a survival benefit to patients with advanced breast cancer with low HER-2 expression. Drug-related hematological toxicity, interstitial pulmonary disease, and other safety issues have also attracted widespread attention. Here, we reviews the researches addressing the efficacy and safety of DS-8201a in the treatment of advanced breast cancer.
-
Keywords:
- antibody–drug conjugates (ADCs) /
- advanced breast cancer /
- DS-8201a
-
目前,抗体药物偶联物(antibody-drug conjugates,ADCs)是抗肿瘤抗体药物研发的新热点,其是由抗体、细胞毒药物和偶联链三部分组成,其抗体通过与肿瘤细胞表面抗原特异性结合,被内化进入靶细胞后,进一步释放所偶联的细胞毒药物而发挥抗肿瘤作用。在2013年,基于既往经紫杉烷(taxane)和曲妥珠单抗治疗失败的人表皮生长因子受体-2(human epidermal growth factor receptor-2,HER-2)阳性转移性乳腺癌患者的总生存期研究数据,曲妥珠单抗-美坦新偶联物T-DM1是第一个获得美国食品和药品监督管理局(FDA)批准用于乳腺癌的ADCs制剂[1]。
T-DM1是由曲妥珠单克隆抗体和微管蛋白抑制剂通过硫脒键偶联组成,可选择性地向肿瘤细胞输送有效载荷的细胞毒药物。由于其膜渗透性低,使得T-DM1对于HER-2低表达肿瘤细胞不能表现出杀伤效应,限制了T-DM1克服HER-2表达异质性的问题,也可能造成后续耐药问题的产生[2],这就促使了新一代基于拓扑异构酶I(topoisomerase I,topo I)抑制剂的ADCs制剂DS-8201a的发展。本文将就DS-8201a在晚期乳腺癌中的研究进展进行综述。
1. DS-8201a的结构与药理学特征
DS-8201a是HER-2靶向抗体药物偶联物,由人源化抗HER-2单克隆抗体、可裂解的四肽链接键和topo I抑制剂伊喜替康甲磺酸盐(DX-8951f)衍生物DXd组成[1]。DXd通过与topo I和DNA复合物形成稳定结合,阻滞DNA的再连接,引起细胞凋亡。DS-8201a的DXd通过马来酰亚胺甘氨酸-苯丙氨酸-甘氨酸(GGFG)四肽链接键与抗体进行连接,该四肽链接键可被组织蛋白酶B和组织蛋白酶L等溶酶体酶裂解,GGFG四肽链接键还可与有效载荷DXd之间进行自降解,使DS-8201a在内化进入肿瘤细胞后能快速水解并释放DXd。
目前,ADCs制剂的药物-抗体比率(drug antibody ratio,DAR)为2~4,T-DM1的DAR为3.5,而DS-8201a为7~8[2]。每个抗体与8个DXd分子的高DAR是传统链间半胱氨酸偶联的最大载荷,更高的DAR可能与不断增加的患者全身不良反应有关。高DAR能够向肿瘤细胞递呈更高浓度的有效药物载荷,也为其具有高效的抗肿瘤活性提供了理论依据。
多项研究显示,在体内和体外实验中DXd具有高度的膜通透性,DS-8201a可通过影响HER-2低表达肿瘤细胞,而表现出“旁观者杀伤效应”,DS-8201a与HER-2阳性肿瘤细胞靶向结合后,释放的DXd分子首先会造成HER-2阳性肿瘤细胞裂解凋亡,随后多个DXd分子会进一步作用于靶向肿瘤细胞旁的HER-2低表达肿瘤细胞,而进一步扩大药物作用范围;在体外实验中,DS-8201a清除了共同培养基上的HER-2阳性和HER-2低表达细胞系,而T-DM1只选择性地影响HER-2阳性细胞系[3-4]。
有研究评估了DS-8201a在人体内的药代动力学特征,其半衰期呈剂量依赖性,中位半衰期为5.7天,血浆中游离DXd浓度较低,与其动物研究结果一致[3]。DXd主要经过细胞色素P4503A4酶进行代谢,有极少量经过葡萄糖醛酸化作用进行代谢。DS-8201a的剂量为3.2~8.0 mg/kg时,血药浓度-时间曲线下面积(area under the curve,AUC)和最大血清药物浓度的增加呈剂量依赖性,依据药代动力学和安全性特征,目前推荐患者使用的DS-8201a剂量为5.4 mg/kg[5-6]。
2. DS-8201a与免疫检查点抑制剂的协同作用
McKenzie等[7]研究表明,topo I抑制剂具有协同免疫检查点抑制剂的作用,如纳武利尤单抗和抗细胞毒性T淋巴细胞抗原抗体,通过激活T细胞活性诱导免疫原性细胞死亡。Iwata等[8]研究发现,在小鼠模型中,使用DS-8201a治疗的小鼠,当再次被接种HER-2阳性和HER-2阴性细胞系,表现为对HER-2阳性和HER-2阴性细胞系有完全和部分的排斥,与未使用过DS-8201a治疗的小鼠不同,后者接种的两种细胞系均可生长,这种效果可能有助于克服由于HER-2表达下调而产生的获得性耐药。该研究为了检测DS-8201a与抗程序性死亡受体-1(programmed cell death protein-1,PD-1)抗体在体内的结合情况,对接种HER-2阳性细胞的小鼠进行单药或联合治疗,结果显示接受联合治疗组的小鼠与单药治疗组相比,具有更佳的存活率和完全缓解率。在DS-8201a和抗细胞毒性T淋巴细胞相关蛋白-4(cytotoxic T-lymphocyte-associated protein 4,CTLA-4)抗体的联合疗法中也观察到相似的结果[9]。
目前,DS-8201a与PD-1抑制剂或抗CTLA-4抗体联合的研究正在进行中,如NCT03523572研究将DS-8201a与纳武单抗联合用于HER-2阳性和HER-2低表达转移性乳腺癌、NCT04042701Ⅰ期临床试验旨在探讨DS-8201a联合派姆单抗用于HER-2阳性和HER-2低表达的晚期乳腺癌[10]。
3. DS-8201a相关的临床研究
3.1 Ⅰ期临床试验
NCT02564900研究是Ⅰ期、公开标签、多中心研究,用以确定DS-8201a的安全性和推荐剂量,研究由两部分组成,第一部分是在日本进行的剂量递增研究,另一部分是在日本和美国进行的剂量扩展研究[11-12]。第一部分入组年龄为20岁以上晚期乳腺癌或胃癌患者,不论HER-2状态,对标准治疗方案已产生耐药,从2015年8月到2016年8月,共24例患者接受剂量为0.8~8.0 mg/kg,每3周1次的DS-8201a静脉输注,研究截止时未发生剂量限制性不良反应或患者死亡。该研究的晚期乳腺癌患者中,客观缓解率(objective response rate,ORR)为41%(7/17),疾病控制率(disease control rate,DCR)为88%(15/17), HER-2阳性患者的治疗反应最佳。另一部分的剂量扩展研究包括5个队列,有115例患者符合研究条件,且未行T-DM1治疗的HER-2阳性晚期乳腺癌患者至少接受1个疗程的DS-8201a治疗,可供评估疗效的111例患者的总有效率为59.5%(66/111)、DCR为93.7%(104/111),中位无进展生存期(progression-free survival,PFS)为22.1个月。NCT02564900研究进一步进行亚组分析显示,之前接受过帕妥珠单抗治疗的乳腺癌患者,总有效率和DCR高于未曾接受过帕妥珠单抗治疗的患者(分别为62.5%和93.8%),中位PFS为16.4个月[13]。
截止至2019年2月,对HER-2低表达的晚期乳腺癌患者进行了评估,共收集54例HER-2低表达的晚期乳腺癌患者数据,大多数为激素受体阳性,已行细胞周期蛋白依赖性激酶4和6(cyclin-dependent kinases 4 and 6,CDK4/6)抑制剂治疗,总缓解率为37%(20/54),所有患者均达到部分缓解,DCR为87%(47/54),中位治疗反应持续时间为10.4个月,中位PFS为11.1个月[14-15]。另有研究的亚组分析显示,47例激素受体阳性患者中,总缓解率为40%(19/47),中位PFS为7.9个月,表明DS-8201a对激素受体阳性晚期乳腺癌患者也有一定疗效[16-17]。2019年12月,美国FDA加速审批了DS-8201a用于治疗不可手术切除或已使用过2种或多种抗HER-2靶向治疗后疾病进展的HER-2阳性晚期乳腺癌[18]。
3.2 Ⅱ期临床试验
DESTINY-Breast 01是一项Ⅱ期多中心研究[19-20],针对正在或已行T-DM1治疗且出现疾病进展的晚期乳腺癌患者,使用DS-8201a剂量为5.4 mg/kg,评估其安全性和有效性。该研究184例接受DS-8201a治疗患者中,有154例为HER-2阳性,28例为HER-2低表达,总缓解率为60.9%(112/184),DCR为97.3%,中位缓解时间(median response duration,MRD)为14.8个月,中位PFS为16.4个月。该研究的亚组分析显示,既往接受过帕妥珠单抗治疗的患者ORR为64%(78/121);激素受体阳性、HER-2阳性的患者ORR为58%(56/97)。DS-8201a对已使用过T-DM1治疗的晚期乳腺癌患者仍然有效。
在DESTINY-Breast 01研究中,伴有中枢神经系统转移患者的亚组分析结果已在2020欧洲肿瘤学会乳腺癌会议上发表,有24例患者在纳入该研究前已出现中枢神经系统转移病灶,ORR为58.3%,包括4.2%完全缓解和54.2%部分缓解,DCR为91.7%,中位PFS为18.1个月[21-23]。
3.3 Ⅲ期临床试验
DESTINY-Breast 02和DESTINY-Breast 03是目前正在进行的Ⅲ期临床试验[22],DESTINY-Breast 02旨在对DS-8201a联合曲妥珠单抗和卡培他滨或联合拉帕替尼和卡培他滨进行比较,用于治疗不可手术切除的晚期乳腺癌或曾使用过T-DM1治疗的转移性乳腺癌;DESTINY-Breast 03旨在对DS-8201a与T-DM1用于曾接受过曲妥珠单抗和紫杉烷治疗且出现疾病进展的HER-2 阳性晚期乳腺癌患者进行对比。另一项DESTINY-Breast 04 Ⅲ期临床试验主要对HER-2 低表达的晚期乳腺癌患者进行研究[24]。
4. 安全性问题
在DESTINY-Breast 01研究中[20],最常见的1级和2级不良反应分别是疲乏(6.0%)和恶心(7.6%)、便秘(5.2%),常见的3、4级不良反应主要是血液学毒性和肺损伤,如中性粒细胞减少(20.7%)、淋巴细胞减少(6.5%)和占9%的间质性肺疾病(interstitial lung disease,ILD),13.6% ILD与DS-8201a相关,而该研究中报道4例死亡病例是由ILD引起。在NCT02564900研究中[12],最常见的3、4级不良反应同样主要是血液学毒性及肺损伤。DS-8201a导致的肺部疾患发生率高于T-DM1,可能与其较高的DAR相关,出现症状的ILD患者应该永久停止使用DS-8201a治疗,并立即进行糖皮质激素剂量≥1 mg/kg治疗,并监测患者的呼吸系统症状[25]。
5. 结语
ADCs药物是一种新型的抗肿瘤治疗方法,随着T-DM1的临床应用加速了该领域的研究,目前除DS-8201a外,SYD985、MEDI4276、XMT-1522等多个药物正在研发。与T-DM1不同,DS-8201a具有更高的药载比、特殊的偶联结构和旁观者杀伤效应,这进一步确立了其在HER-2阳性晚期乳腺癌中的疗效,为晚期乳腺癌患者提供了新的治疗策略。但DS-8201a在HER-2低表达患者中的治疗潜能以及与免疫检查点抑制剂的联合作用、药物不良反应等仍有待进一步研究和证实。
-
[1] Nakada T, Masuda T, Naito H, et al. Novel antibody drug conjugates containing exatecan derivative-based cytotoxic payloads[J]. Bioorg Med Chem Lett, 2016, 26(6):1542-1545. DOI: 10.1016/j.bmcl.2016.02.020
[2] Wu G, Gao Y, Liu D, et al. Study on the heterogeneity of T-DM1 and the analysis of the unconjugated linker structure under a stable conjugation process[J]. ACS Omega, 2019, 4(5):8834-8845. DOI: 10.1021/acsomega.9b00430
[3] Ogitani Y, Hagihara K, Oitate M, et al. 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[J]. Cancer Sci, 2016, 107(7):1039-1046. DOI: 10.1111/cas.12966
[4] Takegawa N, Tsurutani J, Kawakami H, et al. [Fam-]trastuzumab deruxtecan, antitumor activity is dependent on HER2 expression level rather than on HER2 amplification[J]. Int J Cancer, 2019, 145(12):3414-3424.
[5] Lambert JM, Berkenblit A. Antibody-drug conjugates for cancer treatment[J]. Annu Reu Med, 2018, 69:191-207. DOI: 10.1146/annurev-med-061516-121357
[6] Donaghy H. Effects of antibody, drug and linker on the preclinical and clinical toxicities of antibody-drug conjugates[J]. MAbs, 2016, 8:659-671.
[7] McKenzie JA, Mbofung RM, Malu S, et al. The effect of topoisomerase I inhibitors on the efficacy of T-cell-based cancer immunotherapy[J]. J Natl Cancer Inst, 2018, 110(7):777-786. DOI: 10.1093/jnci/djx257
[8] Iwata TN, Ishii C, Ishida S, et al. A HER2-targeting antibody-drug conjugate, trastuzumab deruxtecan (DS-8201a), enhances antitumor immunity in a mouse model[J]. Mol Cancer Ther, 2018, 17(7):1494-1503.
[9] Iwata TN, Sugihara K, Wada T, et al. [Fam-]trastuzumab deruxtecan (DS-8201a) induced antitumor immunity is facilitated by the anti-CTLA-4 antibody in a mouse model[J]. PLoS One, 2019, 14(10):e0222280.
[10] Hurvitz SA, Galsky MD, Shahidi J, et al. A phase Ib, multicenter, open-label study of the antibody-drug conjugate trastuzumab deruxtecan (DS-8201a) combination with nivolumab for advanced HER2-expressing breast or urothelial cancer[J]. Ann Oncol, 2018, 29:121-134.
[11] Nagai Y, Oitate M, Shiozawa H, et al. Comprehensive preclinical pharmacokinetic evaluations of trastuzumab deruxtecan (DS-8201a), a HER2-targeting antibody-drug conjugate, in cynomolgus monkeys[J]. Xenobiotica, 2019, 49(9):1086-1096. DOI: 10.1080/00498254.2018.1531158
[12] 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. DOI: 10.1016/S1470-2045(17)30604-6
[13] Tamura K, Tsurutani J, Takahashi S, et al. Trastuzumab deruxtecan (DS-8201a) in patients with advanced HER2-positive breast cancer previously treated with trastuzumab emtansine: a dose-expansion, phase 1 study[J]. Lancet Oncol, 2019, 20(6):816-826. DOI: 10.1016/S1470-2045(19)30097-X
[14] 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.
[15] Rinnerthaler G, Gampenrieder SP, Greil R. HER2 directed antibody-drug-conjugates beyond T-DM1 in breast cancer[J]. Int J Mol Sci, 2019, 20(5):1115. DOI: 10.3390/ijms20051115
[16] Modi S, Tsurutani J, Tamura K, et al. Trastuzumab deruxtecan (DS-8201a) in subjects with HER2-low expressing breast cancer: updated results of a large phase 1 study[J]. Cancer Res, 2019, 79:617-622.
[17] Montemurro F, Ellis P, Anton A, et al. Safety of trastuzumab emtansine(T-DM1) in patients with HER-2 positive advanced breast cancer: primary results from the KAMILLA study cohort 1[J]. Eur J Cancer, 2019, 109:92-102. DOI: 10.1016/j.ejca.2018.12.022
[18] Li G, Guo J, Shen BQ, et al. Mechanisms of acquired resistance to trastuzumab emtansine in breast cancer cells[J]. Mol Cancer Tber, 2018, 17(7):1441-1453. DOI: 10.1158/1535-7163.MCT-17-0296
[19] 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. DOI: 10.1056/NEJMoa1914510
[20] Krop IE, Saura C, Yamashita T, et al. [Fam-]trastuzumab deruxtecan (T-DXd; DS-8201a) in subjects with HER2-positive metastatic breast cancer previously treated with T-DM1: a phase 2, multicenter, open-label study (DESTINY-Breast01)[J]. Clin Cancer Res, 2020, 80:113-126.
[21] Jerusalem G, Park YH, Yamashita T, et al. CNS metastases in HER2-positive metastatic breast cancer treated with trastuzumab deruxtecan: DESTINY-Breast01 subgroup analyses[J]. Ann Oncol, 2020, 31:1380-1392.
[22] Hunter FW, Barker HR, Lipert B, et al. Mechanism of resistance to trastuzumab emtansine (T-DM1) in HER-2 positive breast cancer[J].Br J Cancer, 2020, 122(5): 603-612.
[23] Angeliki A, Eleni Z, Michalis Li, et al. Trastuzumab deruxtecan (DS-8201a): the latest research and advance in breast cancer[J]. Clin Breast Cancer, 2020, 18:1526-1532.
[24] Modi S, Ohtani S, Lee C, et al. A phase 3, multicenter, randomized, open-label trial of [fam-] trastuzumab deruxtecan (T-DXd; DS-8201a)vs investigator’s choice in HER2-low breast cancer (DESTINY-Breast04)[J]. Clin Cancer Res, 2020, 80:172-183.
[25] Li YJ, Perry SR, Muniz-Medina V, et al. A biparatopic HER2-targeting antibody-drug conjugate induces tumor regression in primary models refractory to or ineligible for HER2-targeted therapy[J]. Cancer cell, 2019, 35(6):948-959. DOI: 10.1016/j.ccell.2019.05.010
-
期刊类型引用(2)
1. 杨爱玲,白丽晓,李佳怡,苏璇,王燕. 德曲妥珠单抗治疗晚期多线治疗失败的乳腺癌的安全性观察. 中国药物应用与监测. 2024(01): 21-24 . 百度学术
2. 白紫合,李娟,吕雅蕾. Her2低表达晚期乳腺癌药物治疗的研究进展. 河北医科大学学报. 2023(07): 860-865 . 百度学术
其他类型引用(3)
计量
- 文章访问数: 346
- HTML全文浏览量: 73
- PDF下载量: 44
- 被引次数: 5