T细胞重定向双特异性抗体在肿瘤治疗中的挑战与应对策略*

秦晓静, 刘雪, 罗文新

中国生物工程杂志 ›› 2023, Vol. 43 ›› Issue (6) : 31-42.

PDF(1469 KB)
PDF(1469 KB)
中国生物工程杂志 ›› 2023, Vol. 43 ›› Issue (6) : 31-42. DOI: 10.13523/j.cb.2302011
综述

T细胞重定向双特异性抗体在肿瘤治疗中的挑战与应对策略*

作者信息 +

Challenges and Therapeutic Strategies for Bispecific T Cell-redirecting Antibodies in Tumor Treatment

Author information +
文章历史 +

摘要

T细胞重定向双特异性抗体能同时结合肿瘤相关抗原和T细胞表面CD3分子,通过将T细胞与肿瘤细胞桥联而激活T细胞发挥抗肿瘤作用,是肿瘤免疫治疗中极具潜力的策略之一。该疗法已成功应用于多种血液肿瘤的治疗,但在实体瘤治疗领域进展缓慢。就近年T细胞重定向双特异性抗体在肿瘤治疗方面所面临的主要挑战及解决策略进行综述,以探讨未来有可能改善其疗效的潜在策略。

Abstract

Bispecific T cell-redirecting antibodies are designed to bind to selected tumor-associated antigens and to CD3 of the T cell receptor. Linking tumor cells and T cells results in activating T cells and inducing targeted T cell-mediated killing of the recognized tumor cells. They have become one of the most promising approaches in tumor immunotherapy. Although this therapy was successfully applied in hematological malignancies therapy, no significant progress in the treatment of solid tumors have been achieved. In this article, we review the major challenges of bispecific T cell-redirecting antibodies, and novel strategies to overcome these hurdles as well as to broaden the indications for this therapy, particularly to solid cancers.

关键词

T细胞重定向双特异性抗体 / 免疫治疗 / 肿瘤

Key words

Bispecific T cell-redirecting antibodies / Immunotherapy / Tumor

引用本文

导出引用
秦晓静, 刘雪, 罗文新. T细胞重定向双特异性抗体在肿瘤治疗中的挑战与应对策略*[J]. 中国生物工程杂志, 2023, 43(6): 31-42 https://doi.org/10.13523/j.cb.2302011
Xiao-jing QIN, Xue LIU, Wen-xin LUO. Challenges and Therapeutic Strategies for Bispecific T Cell-redirecting Antibodies in Tumor Treatment[J]. China Biotechnology, 2023, 43(6): 31-42 https://doi.org/10.13523/j.cb.2302011
中图分类号: Q819   
由于全球环境恶化与人类生活习惯不规律等原因,癌症成为全球主要的公共卫生问题之一,其发病率和死亡率均呈现逐年上升趋势。免疫疗法通过激活自身免疫功能来杀伤肿瘤细胞,在一定程度上能够高质量延长患者生存周期,被认为是肿瘤治疗中极具潜力的策略之一。其中,单克隆抗体药物因具有特异性强、靶向性高、毒副作用低等优点,在肿瘤治疗领域被广泛应用。但是单克隆抗体药物只能单一靶向分子,而肿瘤的发病机制复杂,可能会使肿瘤产生耐药性,甚至脱靶效应[1],在临床上治疗效果往往不够。因此,针对两个及两个以上靶点的双特异性抗体在肿瘤治疗中具有良好前景和发展方向。
双特异性抗体由于含有两种抗体的特异性,可以结合同一种抗原的两种表位或两种不同抗原的表位。双特异性抗体通过效应位点和靶点位点发挥其功能特性。效应位点起到连接募集效应细胞、药物分子或病毒的作用;靶点位点可以靶向到分子或细胞等。T细胞双特异性抗体,尤其是靶向CD3+ T细胞重定向双特异性抗体,成为近年来双特异性抗体的研究热点。抗CD3靶点单抗药物不仅可以通过激活T细胞对肿瘤细胞进行杀伤,而且在一定程度上能够诱导肿瘤细胞凋亡。T细胞表面存在众多CD分子,如CD3、CD4、CD8、CD28等,CD3分子是众多分子中最为关键的T细胞表面分子。CD3分子可以作为诱导T细胞活化的第一信号与T细胞表面的T细胞受体(TCR)组成TCR-CD3复合体,因此在T细胞识别和免疫应答产生过程中显得尤为重要。T细胞重定向双特异性抗体疗法通过将T细胞与肿瘤细胞桥联而发挥作用。这样做,CD3+T细胞的细胞溶解活性可以被重定向到肿瘤细胞,以促进其清除,且不依赖于T细胞与主要组织相容性复合物(MHC)分子,摆脱MHC限制[2]。相比于两种单克隆抗体药物联合使用,T细胞重定向双特异性抗体在很大程度上降低了药物在开发与临床试验中的成本,其将会逐渐成为下一阶段肿瘤免疫治疗中较有前景的靶向药物。
目前有9款双特异性抗体已经获批上市,其中5款药物与CD3靶点相关(表1),超过100多项T细胞重定向双特异性抗体处于临床研究阶段(表2)。Catumaxomab靶向EpCAM和CD3双特异性抗体药物因其制备技术发生在人源化之前,有着很强的免疫原性,同时细胞脱靶后会产生很强的肝毒性,因此在2017年宣布退市。国内重启Catumaxomab双特异性抗体药物针对膀胱癌与胃癌等适应证的研发,正分别处于临床I/Ⅱ期(NCT04799847)和Ⅲ期(NCT04222114)阶段。近年来,T细胞重定向双特异性抗体已经在血液肿瘤治疗领域取得显著成效,并不断拓展到实体瘤治疗领域。2022年6月,靶向CD3和CD20的双特异性抗体Mosunetuzumab经EMA批准上市,用于治疗复发性/难治性(R/R)滤泡性淋巴瘤。目前,该药物也已在国内获批进行临床阶段的研究。同月,康方生物自主研发的用于治疗复发或转移性宫颈癌的PD-1/CTLA-4双特异性抗体药物经国家药品监督管理局(NMPA)批准上市,为肿瘤免疫治疗带来了新的希望。
表1 目前已获批上市的双特异性抗体

Table 1 Bispecific antibodies approved for marketing

双抗药物 靶点 结构/格式 适应证 最早上市时间
Catumaxomab EpCAM×CD3 非对称性IgG like 传统抗体 恶性腹水 2009(EMA)
Blinatumomab CD19×CD3 串联型scFv,不含Fc结构 淋巴细胞白血病 2014(FDA)
Emetazumab factor IXa×
factor X
非对称型IgG like 传统抗体 凝血因子Ⅷ缺乏 2017(FDA)
Amivantamab EGFR×c-Met 非对称型IgG like 传统抗体 血友病 2021(FDA)
Tebentafusp gp100×CD3 串联型scFv,不含Fc结构 不可切除或转移性葡萄膜黑色素瘤 2022(FDA)
Faricimab VEGF×Ang-2 非对称性IgG like 传统抗体 糖尿病黄斑水肿 2022(FDA)
Moseunetuzumab CD20×CD3 非对称型IgG like 传统抗体 复发性/难治性滤泡性淋巴瘤 2022(EMA)
Candonilimab PD-1×CTLA-4 对称型IgG like 传统抗体,
Fc区各串联一个抗CTLA-4的scFv
复发性/转移性宫颈癌 2022(NMPA)
Teclistamab BCMA×CD3 非对称型IgG like 传统抗体 复发性/难治性多发骨髓瘤 2022(FDA)
表2 部分临床在研的T细胞重定向双特异性抗体

Table 2 Representative clinical trials of bispecific T cell-redirecting antibodies

药物名称 靶点 适应证 临床阶段 临床编号 地区
Linvolseltamab BCMA×CD3 多发性骨髓瘤 临床Ⅰ期 NCT05137054 希腊、西班牙
Teclistamab BCMA×CD3 血液恶性肿瘤 临床Ⅰ期 NCT03145181 美国、法国、荷兰、西班牙
Elranatamab BCMA×CD3 多发性骨髓瘤 临床Ⅰ期 NCT05675449 美国
临床Ⅱ期 NCT05090566 美国、加拿大
NCT05228470 中国
NCT05014412 美国、日本、中国、英国
临床Ⅲ期 NCT05020236 阿根廷、澳大利亚、奥地利、比利时、巴西、加拿大、中国、捷克、芬兰、法国、德国、希腊、意大利、日本、韩国、墨西哥、荷兰、新西兰、挪威、波兰、西班牙、瑞典、土耳其、英国
NCT05317416 澳大利亚、奥地利、比利时、加拿大、捷克、芬兰、法国、德国、希腊、匈牙利、印度、以色列、意大利、日本、韩国、荷兰、挪威、波兰、西班牙、瑞典、中国、土耳其
EGFR BAT EGFR×CD3 局部晚期胰腺癌 临床Ⅰ期 NCT03269526 美国
转移性胰腺癌 临床Ⅱ期
ISB 1342 CD38×CD3 复发/难治性多发性骨髓瘤 临床Ⅰ期 NCT03309111 美国、法国
Talquetamab GPRC5D×CD3 复发/难治性多发性骨髓瘤 临床1期 NCT04773522 日本
血液恶性肿瘤 临床Ⅰ期 NCT04634552 美国、比利时、西班牙
临床Ⅰ期 NCT03399799 美国、比利时、荷兰、西班牙
Teclistamab BCMA×CD3 复发/难治性多发性骨髓瘤 临床Ⅰ/Ⅱ期 NCT04696809 日本
血液恶性肿瘤 临床Ⅱ期 NCT04557098 美国
Odronextamab CD20×CD3 B细胞非霍奇金淋巴瘤 临床Ⅱ期 NCT03888105 美国、中国、法国、德国、加拿大、澳大利亚、意大利、日本、韩国、波兰、新加坡、西班牙、英国
REGN5459/
REGN5458
BCMA×CD3 慢性肾病 临床Ⅰ/Ⅱ期 NCT05092347 美国、加拿大、中国、比利时、法国、德国、意大利、荷兰、西班牙、瑞典、英国
Y150 CD38×CD3 复发/难治性多发性骨髓瘤 临床Ⅰ期 NCT05011097 中国
RGV004 CD19×CD3 难治性/复发性B细胞淋巴瘤 临床Ⅰ期 NCT04887025 中国
EMB-07 ROR1×CD3 高级/转移性实体肿瘤 临床Ⅰ期 NCT05607498 澳大利亚、中国
AMG 340 PSMA×CD3 转移性抗阉割前列腺癌 临床Ⅰ期 NCT04740034 美国
TNB-486 CD19×CD3 B细胞非霍奇金淋巴瘤 临床Ⅰ期 NCT04594642 美国、韩国
弥漫性大B细胞淋巴瘤
高级别B细胞淋巴瘤
滤泡性淋巴瘤
GEN1047 B7-H4×CD3 乳腺癌 临床Ⅰ/Ⅱ期 NCT05180474 丹麦、法国、西班牙
子宫癌
卵巢癌
鳞状非小细胞肺癌
Mosunetuzumab CD20×CD3 大B细胞淋巴瘤 临床Ⅱ期 NCT04889716 美国
ARB202 CDH17×CD3 胃肠道癌 临床Ⅰ期 NCT05411133 澳大利亚、中国、新加坡
胆管癌
肝癌
结直肠癌
胰腺癌
胃癌
JNJ-75348780 CD22×CD3 非霍奇金淋巴瘤 临床Ⅰ期 NCT04540796 美国、法国、以色列、韩国、西班牙、中国、英国
慢性淋巴细胞白血病
Odronextamab CD20×CD3 非霍奇金淋巴瘤 临床Ⅰ期 NCT02290951 美国、法国、德国、以色列、英国
慢性淋巴细胞白血病
MGD024 CD123×CD3 急性/慢性骨髓白血病 临床Ⅰ期 NCT05362773 美国
骨髓增生异常综合征
经典霍奇金淋巴瘤
CC-1 PSMA×CD3 前列腺癌复发 临床Ⅰ期 NCT05646550 德国
GB261 CD20×CD3 非霍奇金淋巴瘤 临床Ⅰ/Ⅱ期 NCT04923048 澳大利亚
慢性淋巴细胞白血病
EMB-06 BCMA×CD3 复发/难治性多发性骨髓瘤 临床Ⅰ/Ⅱ期 NCT04735575 澳大利亚、中国
Vibecotamab CD123×CD3 急性髓系白血病 临床Ⅱ期 NCT05285813 美国
骨髓增生异常综合征
Glofitamab CD20×CD3 复发/难治性淋巴瘤 临床Ⅱ期 NCT04703686 法国

1 双特异性抗体的模式结构

双特异性抗体的结构按照功能划分为:(1)含有高变区段的氨基酸序列的可变区抗原结合片段Fab;(2)可与细胞膜上Fc受体结合的的可结晶片段Fc。通常情况下该类抗体含有3个功能区——1个Fc片段和2个Fab片段,结构上呈现左右对称。但IgG4除外,因其铰链区不稳定使Fab段可以随机交换而形成两种抗原结合位点使之成为天然的双特异性抗体。但具有明确特异性的双特异性抗原在自然界中并不多见,大多数需要人工手段进行合成或对天然抗原进行改造。根据不同的设计策略和Fc段的有无进行分类可将其分为两大类:含有Fc片段的双特异性抗体和不含Fc片段的双特异性抗体。

1.1 含有Fc片段的双特异性抗体

(1)三功能抗体(TrioMabs):该类抗体含有3个功能结构域。保留的Fc片段可以招募效应细胞,优先识别辅助细胞上的抗体,两条Fab片段分别识别并结合肿瘤相关抗原和T细胞上的CD3分子。
(2)杵臼结构双特异性抗体(knobs-into-holes):其原理是将两种亲代抗体CH3重链区域进行突变,其中一个由苏氨酸突变为酪氨酸形成杵(knobs)结构,另一个由酪氨酸突变为苏氨酸形成臼(holes)结构,利用空间互补特性将其正确组装并配对。
(3)DVD-Ig结构:在亲代抗体重链与轻链可变区的N端连接另一个抗体的重链与轻链的可变区,形成一个具有双可变、双特异性结合抗原的双特异性抗体。

1.2 不含有Fc片段的双特异性抗体

(1)双特异性抗体T细胞衔接器(BISPECIFIc T cell engaging,BiTE):BiTE由两个不同的单链(single chain Fv,scFv)经一条肽链串联组成,该结构具有两个不同的抗原结合位点,同时抗体的一端可靶向T细胞上的CD3分子。BiTE中的scFv因比较灵活,所以与抗原结合的效率大大提高。
(2)二价的抗体二聚体(diabodies):由两个交叉的scFv构成,重链区域被短肽连接只能以二聚体的形式存在。
其中,因BiTE和Diabodies不含Fc片段与天然抗体结构差别较大,又称为非IgG样类结构。因其结构简单容易大量生产、不含有Fc片段而避免了链交联问题,但也因片段小导致结构不稳定、易聚合失活[1],不含Fc片段导致半衰期短在血浆不宜储存。而含有Fc片段的双特异性抗体(又称类IgG样结构双特异性抗体)可通过突变Fc来调节半衰期[3],且类IgG结构本身在血浆中的半衰期较长,可以长时间稳定保存。

2 双特异性抗体的作用机制——细胞桥联作用

双特异性抗体可以通过介导免疫细胞对肿瘤细胞进行特异性地识别并发挥杀伤作用,这一作用机制可称为细胞桥联作用。双特异性抗体通常以细胞结合器的形式连接两种不同类型的细胞,主要是肿瘤细胞和T/NK细胞。细胞结合器由靶向肿瘤细胞表面相关抗原TAA与结合效应细胞表面抗原的部分组成。现在批准上市的2种CD3+ T细胞重定向双特异性抗体药物catumaxomab和blinatumomab,其发挥作用机制就是重定向T细胞对肿瘤特异性杀伤。CD3分子可以与T细胞表面的T细胞受体(TCR)组成TCR-CD3复合体,无须pMHC与TCR形成复合物,在CD28共刺激作用下作为诱导T细胞活化的第一信号。因此,CD3分子在T细胞识别和免疫应答产生过程中最为常见并尤为关键。
T细胞重定向双特异性抗体一条臂与肿瘤细胞表面抗原TAA结合,另一条臂连接T细胞受体复合物(TCR)中的CD3ε亚基。当靶细胞和效应细胞经CD3+ T细胞结合双特异性抗体比较紧密时,会在两个细胞之间形成免疫突触[4]。T细胞可通过此突触将造孔蛋白(穿孔素)和凋亡诱导蛋白(颗粒酶)转运到靶细胞膜上进行肿瘤细胞的杀伤(图1)。突触的形成还导致TCR交联和T细胞活化,导致促炎症细胞因子释放和T细胞增殖诱导。
图1 双特异性抗体的结构模式

Fig.1 Formats of different bispecific antibodies

Full size|PPT slide

图2 T细胞重定向双特异性抗体的作用机制

Fig.2 Mechanism of action of T cell redirection bispecific antibodies

Full size|PPT slide

3 T细胞重定向双特异性抗体在实体瘤中的挑战

3.1 细胞因子释放综合征

细胞因子释放综合征(CRS)被认为是由免疫细胞过度激活导致的,是一种严重的免疫副反应[5]。其特征是:血清中IL-6、IL-10、肿瘤坏死因子-ɑ(TNF-ɑ)和γ干扰素(IFN-γ)等促炎症细胞因子急剧升高,进而导致细胞因子风暴发生。CRS的临床表现包括发热、疲惫、肌肉疼痛、呼吸窘迫、毛细血管渗漏、血管舒张性休克和最终的系统性器官衰竭[6]。作为首款上市双特异性抗体药物Catumaxomab,虽然在治疗恶性腹水中展现出了良好疗效,但因其有着很强的免疫原性,细胞脱靶后会产生很强的肝毒性,同时引发强烈的细胞因子风暴[7],因此于2017年宣布退市。2022年8月,一项用于评估BRG1/BRM抑制剂FHD-286在治疗复发/难治性急性髓细胞性白血病(AML)和骨髓增生异常综合征(MDA)的临床试验被FDA暂停,原因是出现一名受试者死亡的严重不良反应事件(NCT04891757)。初步认为是由于试验药物FHD-286(靶向BRG1×BRM)靶向机制所引起的。给药后观察到受试者体内白细胞增多,大量的细胞因子IL-1、IL-6、IL-8等被快速释放。

3.2 靶向性与非靶向性的细胞毒性

T细胞重定向双特异性抗体免疫治疗面临的第二个挑战是如何特异性识别并区分正常细胞与肿瘤细胞的治疗靶点。正在开发的抗体药物大多是分化标志物[8],目前在研大多数靶向实体瘤的靶点,如CEA、EGFR、EpCAM、HER2等都属于分化标志物范畴[9]。这类物质的特点是可以在正常细胞中表达,但表达水平受限。在一定程度上缩小治疗窗口,限制T细胞重定向双特异性抗体的设计和开发。因此,如何使抗体选择性结合靶点高表达的肿瘤,而不结合低表达的正常细胞是CD3双特异抗体面临的一个难题。

3.3 肿瘤微环境的T细胞浸润不足及反向招募T细胞亚群

实体肿瘤与恶性血液肿瘤的一个根本区分特征是实体瘤区域存在免疫抑制肿瘤微环境(tumor microenvironment, TME)。TME主要由肿瘤免疫抑制细胞、基质细胞及免疫抑制细胞因子和一些可溶性分子构成,它们都具有抑制抗肿瘤免疫的功能[10]。肿瘤细胞和免疫抑制细胞中均存在抑制免疫检查点分子,抑制免疫检查点分子的上调往往会导致肿瘤细胞免疫逃逸。此外,肿瘤细胞还存在其他免疫逃避机制。正常情况下,免疫激活分子和免疫抑制分子之间存在动态平衡,共同维持着机体免疫系统的稳定性。但当肿瘤细胞或T细胞表面的免疫抑制因子过度表达后会造成T细胞功能紊乱与耗竭,导致肿瘤微环境中的CD8+T细胞浸润不足。肿瘤浸润淋巴细胞(tumors infiltrate lymphocytes, TIL)不足还有可能是因为肿瘤微环境中的T细胞长期处于抗原刺激之下,肿瘤特异性T细胞克隆化频率下降,效应T细胞不断减少,从而导致T细胞耗竭。
T细胞重定向双特异性抗体必须与肿瘤靶细胞和效应T细胞接触,短暂的接触可以促进T细胞与肿瘤细胞之间形成免疫突触。之后会在免疫突触观察到穿孔素和颗粒酶的存在,经T细胞导向肿瘤细胞进行杀伤。大部分细胞毒性T细胞(cytotoxic T cells, CTL)都可以通过这种方式参与肿瘤细胞的裂解,如CD8+ T细胞、CD4+ T细胞、γδT细胞、NKT细胞,通常情况下CTL中的CD8+ T细胞为主要效应细胞。将T细胞重定向双特异性抗体药物静脉注射到体内,发现双特异性抗体可直接招募或激活CTL细胞,也可以招募其他免疫细胞。其可无差别招募多种T细胞,尤其是对一些起反作用的CD3+ T细胞亚群的招募,包括原始或衰竭T细胞、调节性T细胞和辅助性T细胞亚群(helper T cells, Th),产生耐药性,削弱其杀伤肿瘤细胞的疗效。一项临床研究表明,Binatumomab的耐药性是由CD3+ CD4+ CD25 hiFoxP3+ Treg细胞导致的[11],从而阻碍CD8+T细胞溶解,抑制CD8+ T细胞肿瘤杀伤作用。除此之外,招募到的CD4+ T细胞主要是Th1、Th17,往往会释放一些细胞因子而引起细胞因子风暴。因此提高双特异性抗体的选择性,可以更好地进行细胞杀伤及降低细胞毒性作用。除此之外,寻找其他T细胞招募或激活靶点如CD28也是解决T细胞在免疫治疗障碍中T细胞结合双特异性抗体无差别的招募应对策略之一。

4 T细胞重定向双特异性抗体的技术优化和治疗策略开发

4.1 调动其他免疫细胞对肿瘤细胞进行定向杀伤与清除

细胞衔接器连接肿瘤细胞和免疫细胞形成免疫突触介导杀伤作用。免疫细胞可以是T细胞也可以是自然杀伤(natural killer, NK)细胞。双特异性抗体T细胞衔接器(BiTE)通过针对TAA和CD3抗体将肿瘤细胞与T细胞结合,以介导T细胞杀伤肿瘤细胞。双特异性抗体NK细胞衔接器(bispecific killer cell engager, BiKE)将CD16A+ NK细胞重定向到肿瘤细胞,并诱导NK细胞活化。与BiTE相比,BiKE具有减缓细胞因子释放综合征和降低神经毒性的疗效。AFM13是首款靶向CD30和CD16的四价双特异性抗体,可以同时结合NK细胞上的CD16A和淋巴瘤细胞上的CD30,从而介导NK细胞对癌细胞的杀伤与清除[12]。使用改良免疫细胞(AFM13-NK)和双特异性抗体(AFM13)治疗复发性或难治性CD30+霍奇金或非霍奇金淋巴瘤患者的临床试验正处于I/Ⅱ期阶段(NCT04074746)。使用AFM13免疫治疗可能有利于患者的免疫系统攻击肿瘤细胞,并可能阻碍肿瘤细胞生长和转移。与NK细胞连用的AFM13比单独使用AFM13本身可能会杀死更多肿瘤细胞,并抑制肿瘤生长。

4.2 开发不同格式的T细胞重定向双特异性抗体

迄今为止,约九成双特异性抗体用于肿瘤治疗[13]。在肿瘤治疗领域,双特异性抗体的半衰期随其分子大小而变化。对于一些分子量小的双特异性抗体,如缺少Fc片段的双特异性T细胞衔接器(BiTE),仅通过抗原结合力发挥治疗作用,免疫原性较低,给药频率次数增多,有必要延长其半衰期。使用多价态靶向CD3的T细胞双特异性抗体进行肿瘤细胞杀伤,其结果显示比一加一具有更好的效果。靶向抗原的不同位置和不同的亲和力也会导致效果的巨大差异[14]。双特异性抗体中提高CD3亲和力是非必需的,因为CD3的低亲和力BsAb在体内仍显示出相同的杀伤效果,并且比CD3高亲和力的双特异性抗体(CD3×CLL1)具有更好的安全性和良好的耐受性[15]。因此,双特异性抗体通常使用CD3的低亲和力抗体和TAA的高亲和力抗体[16-19]。不同表达水平的TAAs需要选择具有不同亲和力的抗体。不同结构格式具有不同效果。例如,与基于串联型scFv的双特异性抗体相比,基于Fab的双特异抗体具有更好的生物物理性质。此外,基于Fab的双特异性抗体不仅保留了其串联型scFv对应物的活性,而且具有独特的生物活性[20]。针对多发性骨髓瘤的治疗,许多生物公司聚焦在一种靶向T细胞表面抗原CD3和B细胞成熟抗原BCMA双特异性抗体上。Elranatamab是靶向BCMA和CD3双特异性抗体药物,其对BCMA和CD3结合亲和力进行了优化。当抗体亲和力降低时,更有利于激活T细胞,增强T细胞抗骨髓瘤的活性,在一定程度上减少了非靶向的细胞毒性。在其他临床试验研究中,研究人员设计开发了5种不同格式的T细胞重定向双特异性抗体(均靶向BCMA×CD3),使用具有不同稀释度的BCMA×CD3双特异性抗体与CHO-BCMA靶细胞和T细胞共孵育,实时检测细胞毒性,所有设计的带有T细胞的BCMA×CD3双特异性抗体存在剂量依赖性[21]。单独的BCMA×CD3双特异性抗体没有T细胞的细胞毒性活性。同样靶向BCMA×CD3双特异性抗体TNB-383B具有强大的T细胞激活能力,同时还能降低对CD3分子的亲和力。TNB-383B的I期临床研究表示,试验者接受的剂量在0.025~1.8 mg范围内,随着剂量的增加反应率不断上升。后续剂量上升至40 mg左右,试验者的肿瘤体积明显得到缓解,客观缓解率为80%(NCT03933735)。
双特异性抗体构型的设计会影响每个靶标的结合位点数量。每个靶标具有一个结合位点的二价双特异性抗体表示为1+1,加入其他结合位点可导致三特异性抗体(2+1)甚至是四特异性抗体(2+2或1+3)的产生。尽管传统的双特异性抗体以1:1的CD3和TAA价对称设计,但通过1+2的设计增加结合能力可能会增强对肿瘤细胞的识别,同时避免在缺乏TAA的情况下对T细胞上CD3的激活。在靶点数目上,主要以双特异性抗体为主。针对三特异性抗体的设计多是在双特异性抗体的基础上进行设计与改造。Seung等[22]在HER2×CD3双特异性抗体的基础上在抗CD3的可变区域偶联上一个抗CD28的可变区结构域,针对HER2/CD3×CD28的三特异性抗体在人源化小鼠模型中通过CD4+ T细胞和CD8+ T细胞介导乳腺癌细胞消退。三特异性抗体中使用的单价抗CD28不会诱导先前使用二价超级拮抗剂抗CD28时出现的严重炎性细胞因子释放[23]。尽管HER2在肿瘤中过度表达可能会增加全身炎症,但它可以诱导局部炎性细胞因子和趋化因子释放,这有助于激活周围的免疫细胞,也有助于免疫细胞运输到肿瘤。三特异性抗体的多价数可作为一种理想的抗肿瘤机制,作为一种具有肿瘤免疫治疗前景的多靶向免疫干预。
除了降低抗体亲和力和改变抗体价数外,还可以利用前药(pro-drug)设计概念来降低双特异性抗体药物对正常组织细胞的特异性杀伤,大大提高双特异性抗体药物的有效性与安全性。肿瘤细胞和正常组织细胞差异较大的是一些胞内蛋白,只能通过HLA I类分子进行呈递,因此常规抗体无法被递送至胞内发挥功效。Immunocore公司开发设计了一类TCR双特异性融合蛋白——Tebentafusp,可由肽-MHC(pMHC)复合物呈现,似乎是我们能够确定的最接近的“理想靶点”。Tebentafusp是一种ImmTAC分子,靶向gp100×CD3。ImmTAC分子由TCR靶向结构域与单链可变片段(scFv)抗CD3效应结构域组成。TCR靶向结构域结合细胞表面作为人类白细胞抗原(HLA)-肽(pHLA)复合物呈现的肽,抗CD3结构域参与并激活CD3+ T细胞,可将CD3+ T细胞重新定向到表达gp100的黑色素瘤细胞中,诱导肿瘤细胞溶解[24]。细胞裂解后,Tebentafusp可使凋亡肿瘤细胞释放的肿瘤相关抗原,而后被树突状细胞捕获激活后续的免疫应答。
另有一种应对靶向性与非靶向性细胞毒性的解决策略是选择性激活。在正常组织中,双特异性抗体药物以前体形式存在,没有活性(图3)。在到达肿瘤区域特定部位后,在某些酶的作用下进行酶切,去除遮蔽表位的一小段多肽,暴露结合位点,行使双特异性抗体的功能[25]。ADG138是靶向HER2×CD3的双特异性抗体,研究者对HER2和CD3的表位进行遮蔽处理。到达特定部位后,遮蔽肽(masking peptide)被去除,露出抗体的Fab区域,暴露抗HER2和抗CD3结合位点,与肿瘤细胞上的抗原进行正常结合,招募并激活T细胞对肿瘤细胞进行特异性杀伤。
图3 前体双特异性抗体的结构示意图及其作用原理

Fig.3 Design of probody and functional mechanism

Full size|PPT slide

4.3 降低CRS的发生频率与风险

临床试验中,大多是使用IL-6、TNF-ɑ相关的抗体药物来预防和控制细胞因子释放综合征。Blinatumomab临床试验的血液学和实体瘤指征表示,使用T细胞重定向双特异性抗体(靶向CD19×CD3)治疗急性淋巴白血病患者后,在患者体内均可观察到细胞因子释放综合征,其严重程度取决于治疗类型和靶点[26-27]。使用人源化小鼠模型进行临床前研究,发现T细胞重定向双特异性抗体诱导的细胞因子释放综合征的主要媒介分子是活化T细胞产生的TNF-ɑ,进而导致单核细胞大量分泌炎性细胞因子[28]。阻断上游的TNF-ɑ和下游的IL-1β或IL-6可以缓解细胞因子释放综合征[29]。也有研究报告表明,皮下注射T细胞重定向双特异性抗体或递增剂量可以降低细胞因子释放综合征的程度[30-31]。在临床前模型C57BL/6小鼠中,分别将GD2导向三特异性抗体药物Surek(anti-GD2×anti-murine CD3)进行皮下给药和静脉输送。在注射两次Surek时,皮下和静脉注射表现出相同的抗肿瘤效果。但进行第三次注射后,皮下给药的生存率可达85%,而静脉注射组因抗体药物聚集所带来的严重不良反应必须立即执行安乐死。在小鼠和食蟹猴模型中进行的多项实验研究表明,降低CD3亲和力可以降低治疗诱导的细胞因子水平[,32-34]
临床上已经测试了一种启动剂量策略,以减轻细胞因子释放综合征引起的毒性。该方法包括先给较低的初始药物剂量,然后给予较高的维持剂量(NCT02152956),随后引入了定量细胞因子模型,以改进临床给药策略的设计,从而在最小化细胞因子释放综合征相关毒性的同时实现疗效。2019年辉瑞生物公司研发出新的药代动力学-药效学(PK-PD)模型模拟了T细胞导向疗法治疗后的细胞因子释放情况[35]。随后建立了一个定量系统药理学模型,以准确预测与生物标志物相关的安全性和有效性,从而降低抗CD3×抗CD20细胞因子风暴的风险。另外,针对非霍奇金淋巴瘤中的双特异性抗体Mosunetuzumab(NCT02500407),后续针对之前接受过两次或两次以上治疗的复发或难治性滤泡性淋巴瘤患者的安全性和抗肿瘤活性进行评估,发现固定持续时间的Mosunetuzumab具有良好安全性,对细胞因子释放综合征具有较高的缓解率[36]
T细胞重定向双特异性抗体的效力和安全性受到多种因素的影响,其中CD3结合的亲和力决定着双特异性抗体的疗效和潜在毒性。再生元研究团队将Jurkat/NFAT-luc细胞与表达人MUC16的OVCAR-3细胞、表达人PSMA的C4-2细胞、表达人CD20的RAJI-KO/CD80/CD86细胞、表达人BCMA的H929细胞与连续稀释的抗原MUC16、PSMA、CD20、BCMAxCD3双特异性抗体或CD3双特异性对照抗体一起孵育[37],结果显示,中等亲和力的双特异性抗体的杀伤作用高于弱亲和力和微弱亲和力的。但是亲和力高的双特异性抗体并不代表其杀伤作用好,更高亲和力的T细胞重定向双特异性抗体有着更高水平的T细胞激活,导致更高强度的细胞杀伤作用。研究人员基于对具有较弱亲和力CD3双特异性抗体的研究,发现CD3臂结合亲和力降低仍然可以做出有效的抗肿瘤反应,同时限制全身细胞因子水平,并在肿瘤模型中触发了较低水平的细胞因子。

4.4 联合治疗策略

4.4.1 不同靶点双特异性抗体联合放射性治疗技术

放射性治疗是指通过放射线来治疗肿瘤细胞,已成为治疗恶性肿瘤最主要的手段之一。将肿瘤靶向药物(双特异性抗体)与有效载荷(放射性同位素)结合,分多步进行联合靶向免疫治疗。双特异性抗体给药24 h后,给予自由基清除剂,在4 h后注射放射性金属,可以弥补传统放射性治疗中放射性同位素被肿瘤摄取不足、在肾脏中保留率高的不足。有文章报道一种自组装和拆卸(self-assembling and disassembling, SADA)双特异性抗体,与p53蛋白融合形成p53-SADA-BsAbs四聚体。给药后在血液中循环形成二聚体或单体,经肾脏过滤后可以清除、降低大鼠免疫原性,并且对骨髓、肾脏或肝脏没有任何短期或长期毒性[38]。在使用抗GD2双特异性抗体的预靶向放射性免疫治疗神经母细胞瘤时,增加放射性治疗指数发现具有缩小肿瘤、消除微转移以及防止复发的疗效。
放射性治疗通过刺激免疫系统发挥作用,增加了肿瘤浸润免疫刺激细胞的丰度,增强了肿瘤细胞的免疫原性,提高了抗肿瘤免疫能力。但也会增加患者局部或全身免疫抑制,在较低剂量时仍会造成骨髓暂时性功能消退,最终导致淋巴细胞减少。放射性治疗在一定程度上会损伤CD8+ T细胞,而免疫治疗的应答依赖于CD8+ T细胞的激活。因此,在某些情况下放射性治疗不能增强与免疫治疗的联合治疗效果,有时会削弱免疫治疗效果。

4.4.2 双特异性抗体联合单克隆抗体的治疗策略

双特异性抗体与单克隆抗体联合的治疗策略大多处于临床前阶段或者临床阶段。早在2020年,针对HLA-A*02:01阳性MAGE-A4阳性晚期实体肿瘤患者临床阶段的治疗就首次采用了抗TCR/CD3双特异性抗体联合Atezolizumab单克隆抗体的治疗策略(NCT03973333)。2021年,康方生物宣布抗TIGIT单克隆抗体AK127与靶向PD-1/CTL-4双特异性抗体AK104联合治疗晚期或转移性实体瘤的方案进入临床I期[39]。临床前期试验结果显示AK104双特异性抗体联合AK127单抗在小鼠模型中具有显著的抗肿瘤效果。AK127单抗药物阻止AK127(抗TIGIT单抗)与其配体CD155结合,阻碍了T/NK细胞攻击肿瘤细胞的抑制信号。AK127与TIGIT特异性结合,抑制TIGIT与其配体之间的相互作用,免疫抑制得到缓解,促进抗肿瘤作用。TIGIT表达与PD-1密切相关,尤其是在肿瘤浸润性T细胞中,这2个靶点通常在同一细胞上共表达,这为开发抗TIGIT药物联合抗PD-1抗体提供了科学依据。目前,AK104双特异性抗体与单抗AK117(CD47单抗)、AK109(VEGFR单抗)、AK119(CD73单抗)联合治疗策略正在逐步推进。

4.4.3 双特异性抗体联合免疫抑制剂的治疗策略

免疫抑制剂是一类对机体免疫反应具有抑制作用的药物,通常被用于器官移植免疫排斥反应和自身免疫病。当双特异性抗体靶向肿瘤组织部位时,因对其抗原表位的亲和力较强可能会引发较强的免疫原性,诱发细胞因子快速而剧烈地释放,引发细胞因子风暴。免疫抑制剂与双特异性抗体联用在一定程度上可以避免T细胞过度活化,提高药物的安全性。
FTY720(Fingolimod)是一类经FDA批准用于治疗慢性淋巴细胞白血病和套细胞淋巴瘤(MCL)的免疫抑制剂。有研究发现FTY720可以增加CD74表达,并使MCL细胞敏化为米拉妥珠单抗(抗CD20人源化抗体)介导的细胞死亡[40],因此设计并构建抗CD20/CD74双特异性抗体与FTY720联用进行抗肿瘤联合治疗。与单独使用FTY720、抗CD20单克隆抗体或抗CD47单克隆抗体相比,抗CD20/CD74双特异性抗体与FTY720联合治疗有21%的活细胞,说明两种制剂存在协同作用。CEA/CD3 T细胞结合双特异性抗体是一种新型的T细胞双特异性抗体,靶向肿瘤细胞上的CEA和T细胞上的CD3。在临床前,CEA-CD3 T细胞结合双特异性抗体具有强大的抗肿瘤活性,导致肿瘤内T细胞浸润和激活增加、T细胞介导的肿瘤细胞杀戮和PD-L1/PD-1上调[18]。将靶向CEA和CD3的T细胞结合双特异性抗体单独治疗转移性结肠癌与Atezolizumab免疫抑制剂联合治疗的效果进行比较,结果表明联合治疗策略具有更强的抗肿瘤活性与生物安全性。

4.5 其他治疗策略

溶瘤病毒具有感染、复制和溶解肿瘤细胞的能力,在增强免疫治疗的同时诱导免疫原性肿瘤微环境渗透性增强,使CD8+ T细胞在肿瘤微环境中得到充分浸润[41]。溶瘤病毒作为单一疗法因正常细胞摄取导致其非靶向性地溶解细胞,因此临床试验中多采用溶瘤病毒联合治疗策略,并取得较大进展[42]。Yu 等[43]构建了一种可携带肿瘤细胞表面抗原EphA2的BiTE抗体的溶瘤牛痘病毒(VV),将其命名为EphA2 TEA VV。在肺癌异种移植小鼠模型中,EphA2 TEA VV可激活和重定向细胞毒性T细胞(CTL)的浸润,比常规溶瘤牛痘病毒展现出更有效的抗肿瘤作用。在另一项研究中,Wang等[44]设计了一种可搭载MUC16(人黏蛋白16)的BiTE抗体的溶瘤腺病毒OAd-MUC16-BiTE。局部给药后,BiTE在肿瘤局部组织中可持续稳定表达,可降低全身给药引起的毒性,其介导的T细胞活化和靶细胞裂解具有抗原特异性。此外,OAd-MUC16-BiTE可“逆转”肿瘤微环境,促进CTL浸润,增强T细胞介导的肿瘤细胞的杀伤效应,显著提高了抗肿瘤活性。以上研究均表明溶瘤病毒和BiTE联合使用所获得的协同效应突破了单独给药的局限性。
将BiTE与免疫检查点抑制剂联合应用也具有增强抗肿瘤的效果。为了克服单独使用BiTE治疗恶性肿瘤的难治性,联合使用BiTE与抗PD-1激动剂和抗4-1BB激动剂,肿瘤微环境中CD8+T细胞活性增强,展现出很强的抗肿瘤特性[45]。一项在研临床试验中,研究者将PD-1免疫检查点抑制剂Pembroblizumab与BiTE抗体Blinatumomab联合用于复发/难治性急性白血病或淋巴瘤患者的治疗,前期安全性和可行性得到验证,但因其缺乏注册对复发/难治性急性白血病提供新的抢救疗法而被暂停临床I期试验(NCT03605589)[46]。需要指出的是,BiTE因其Fc段缺失导致半衰期缩短,需要寻找更加有效的结构模式进行改造以提高其在体内的半衰期。

5 总结与展望

抗体药物尤其是以CD3为代表的T细胞重定向双特异性抗体在肿瘤治疗领域取得了革命性突破,其已成功应用于多种血液肿瘤的治疗,但在实体瘤治疗领域进展缓慢,仍然面临着诸多严峻挑战和待优化之处,包括引发细胞因子综合征、靶向性与非靶向性的细胞毒性、肿瘤微环境中T细胞浸润较低及肿瘤微环境中T细胞的活性被抑制等(表3)。细胞因子释放综合征是一种严重的免疫反应,已上市的Binatumomab单抗和两款CAR-T药物(Kymriah和Yescarta)在临床使用中均会引发细胞因子风暴的毒副作用。现有预防和治疗细胞因子释放综合征的主要手段是使用IL-6和TNF-α相关的抗体药物。靶向性与非靶向性的细胞毒性、效应细胞数量有限以及肿瘤中T细胞活性被抑制这3个挑战之间相互制约,因此解决T细胞重定向双特异性抗体弊端时三者缺一不可。现阶段,随着科学技术的不断发展,为更大程度地发挥T细胞重定向双特异性抗体作用,研究者提出了将其与其他策略联合使用,如与免疫检查点抑制联合使用、与免疫激活剂联合使用、与溶瘤病毒联合使用等,这些策略的提出为肿瘤治愈增添了可能。此外,由于靶向单一抗原可能会造成肿瘤抗原丢失,为增强抗体对肿瘤细胞的靶向性,也为了降低耐药性,可采用同时靶向两个不同肿瘤抗原,开发可靶向激活T细胞的三特异性抗体的策略来解决。相信随着免疫治疗技术的不断发展,人类终将攻克肿瘤这一难题。
表3 T细胞重定向双特异性抗体面临的挑战与解决策略

Table 3 Challenges and therapeutic strategies for bispecific T cell-redirecting antibodies

双特异性抗体面临的挑战 特征 解决策略
细胞因子释放综合征 IL-6、IL-10、TNF-ɑ、IFN-γ水平急剧升高,引发细胞因子风暴 IL-6和TNF-α抗体药物,免疫抑制剂等
反向招募T细胞亚群 无差别招募原始/衰竭T细胞、CD4+ T细胞、调节性T细胞等 提高双特异性抗体的选择性
靶向性与非靶向性的细胞毒性 正常细胞低水平抗原表达导致有害的靶向肿瘤毒性 前药、TCR融合蛋白+抗体双功能抗体、“2+1”格式CD3双抗;双抗体内分布重排等
肿瘤微环境 免疫检查位点 T细胞功能障碍和衰竭导致抗肿瘤免疫抑制 联合免疫共抑制或免疫共刺激
免疫抑制细胞与免疫
抑制细胞因子
免疫细胞、细胞因子有助于抑制抗肿瘤免疫反应 溶瘤病毒
T细胞耗竭、活性限制 CD3+ T细胞重定向受阻 双肿瘤抗原靶向CD3+ T细胞导向剂的研制

参考文献

[1]
Wu S Y, Wu F G, Chen X Y. Antibody-incorporated nanomedicines for cancer therapy. Advanced Materials, 2022, 34(24): 2109210.
[2]
Deshaies R J. Multispecific drugs herald a new era of biopharmaceutical innovation. Nature, 2020, 580(7803): 329-338.
[3]
Acheampong D O. Bispecific antibody (bsAb) construct formats and their application in cancer therapy. Protein & Peptide Letters, 2019, 26(7): 479-493.
[4]
Xiong W, Chen Y H, Kang X, et al. Immunological synapse predicts effectiveness of chimeric antigen receptor cells. Molecular Therapy, 2018, 26(4): 963-975.
Chimeric antigen receptor (CAR)-modified T cell therapy has the potential to improve the overall survival of patients with malignancies by enhancing the effectiveness of CAR T cells. Precisely predicting the effectiveness of various CAR T cells represents one of today's key unsolved problems in immunotherapy. Here, we predict the effectiveness of CAR-modified cells by evaluating the quality of the CAR-mediated immunological synapse (IS) by quantitation of F-actin, clustering of tumor antigen, polarization of lytic granules (LGs), and distribution of key signaling molecules within the IS. Long-term killing capability, but not secretion of conventional cytokines or standard 4-hr cytotoxicity, correlates positively with the quality of the IS in two different CAR T cells that share identical antigen specificity. Xenograft model data confirm that the quality of the IS in vitro correlates positively with performance of CAR-modified immune cells in vivo. Therefore, we propose that the quality of the IS predicts the effectiveness of CAR-modified immune cells, which provides a novel strategy to guide CAR therapy.Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.
[5]
Morris E C, Neelapu S S, Giavridis T, et al. Cytokine release syndrome and associated neurotoxicity in cancer immunotherapy. Nature Reviews Immunology, 2022, 22(2): 85-96.
[6]
Frey N. Cytokine release syndrome: who is at risk and how to treat. Best Practice & Research Clinical Haematology, 2017, 30(4): 336-340.
[7]
Frampton J E. Catumaxomab. Drugs, 2012, 72(10): 1399-1410.
Catumaxomab is a rat/murine hybrid, trifunctional, bispecific (anti-human epithelial cell adhesion molecule [EpCAM] × anti-CD3) monoclonal antibody. Compared with paracentesis alone, paracentesis followed by catumaxomab therapy was associated with significant prolongation of paracentesis-free survival and time to repeat paracentesis in a randomized, open-label, multicentre, pivotal phase II/III trial in patients with recurrent symptomatic malignant ascites due to EpCAM-positive tumours who were resistant to conventional chemotherapy. The benefits of catumaxomab were seen across a broad range of epithelial ovarian and nonovarian cancers, and irrespective of whether or not catumaxomab recipients developed human anti-mouse antibodies. Combining catumaxomab with paracentesis also resulted in more pronounced and prolonged reductions in ascites signs and symptoms and a delayed deterioration in health-related quality of life compared with paracentesis alone. Despite the study not being designed or powered to evaluate overall survival, significant differences favouring the addition of catumaxomab to paracentesis were seen in analyses of the safety population and the subpopulation of patients with gastric cancer. Catumaxomab was generally well tolerated in the pivotal phase II/III trial. The most frequent adverse events attributed to catumaxomab treatment included cytokine-release-related symptoms, which were mostly of mild to moderate severity and manageable with standard symptomatic treatment.
[8]
Wu Z, Cheung N V. T cell engaging bispecific antibody (T-BsAb): from technology to therapeutics. Pharmacology & Therapeutics, 2018, 182: 161-175.
[9]
Peters I T A, Hilders C G J M, Sier C F M, et al. Identification of cell-surface markers for detecting breast cancer cells in ovarian tissue. Archives of Gynecology and Obstetrics, 2016, 294(2): 385-393.
The safety of ovarian tissue autotransplantation in oncology patients cannot be ensured, as current tumor-detection methods compromise the ovarian tissue viability. Although non-destructive methods (for instance near-infrared fluorescence imaging) can discriminate malignant from healthy tissues while leaving the examined tissues unaffected, they require specific cell-surface tumor markers. We determined which tumor markers are suitable targets for tumor-specific imaging to exclude the presence of breast cancer cells in ovarian tissue.Immunohistochemistry was performed on formalin-fixed, paraffin-embedded specimens of ten ovaries from premenopausal patients. Additionally, we screened a tissue microarray containing tumor tissue cores from 24 breast cancer patients being eligible for ovarian tissue cryopreservation. The following cell-surface tumor markers were tested: E-cadherin, EMA (epithelial membrane antigen), Her2/neu (human epidermal growth factor receptor type 2), αvβ6 integrin, EpCAM (epithelial cell adhesion molecule), CEA (carcinoembryonic antigen), FR-α (folate receptor-alpha), and uPAR (urokinase-type plasminogen activator receptor). For each tumor, the percentage of positive breast tumor cells was measured.None of the ten ovaries were positive for any of the markers tested. However, all markers (except CEA and uPAR) were present on epithelial cells of inclusion cysts. E-cadherin was present in the majority of breast tumors: ≥90 % of tumor cells were positive for E-cadherin in 17 out of 24 tumors, and 100 % of tumor cells were positive in 5 out of 24 tumors.Of the markers tested, E-cadherin is the most suitable marker for a tumor-specific probe in ovarian tissue. Methods are required to distinguish inclusion cysts from breast tumor cells.
[10]
Tian Z, Liu M, Zhang Y, et al. Bispecific T cell engagers: an emerging therapy for management of hematologic malignancies. Journal of Hematology & Oncology, 2021, 14(1): 1-18.
[11]
Singh A, Dees S, Grewal I S. Overcoming the challenges associated with CD3+ T-cell redirection in cancer. British Journal of Cancer, 2021, 124(6): 1037-1048.
The development of bispecific antibodies that redirect the cytotoxic activity of CD3+ T cells to tumours is a promising immunotherapeutic strategy for the treatment of haematological malignancies and solid cancers. Since the landmark FDA approval at the end of 2014 of the anti-CD3 × anti-CD19 bispecific antibody blinatumomab (Blincyto) for the treatment of relapsed/refractory B-cell acute lymphoblastic leukaemia, ~100 clinical trials investigating the safety and efficacy of CD3+ bispecific T-cell redirectors for cancer have been initiated. However, despite early success, numerous challenges pertaining to CD3+ T-cell redirection in the context of cancer exist, including the recruitment of counterproductive CD3+ T-cell subsets, the release of systemic cytokines, the expansion of immune checkpoint molecules, the presence of an immunosuppressive tumour microenvironment, tumour antigen loss/escape, on-target off-tumour toxicity and suboptimal potency. The aim of the present review is to discuss novel approaches to overcome the key challenges associated with CD3+ bispecific T-cell redirection in order to achieve an optimal balance of anti-tumour activity and safety.
[12]
Kerbauy L N, Marin N D, Kaplan M, et al. Combining AFM13, a bispecific CD30/CD16 antibody, with cytokine-activated blood and cord blood-derived NK cells facilitates CAR-like responses against CD30+ malignancies. Clin Cancer Res, 2021, 27(13): 3744-3756.
Natural killer (NK) cell recognition and function against NK-resistant cancers remains substantial barriers to the broad application of NK cell immunotherapy. Potential solutions include bispecific engagers that target NK cell activity via an NK activating receptor when simultaneously targeting a tumor-specific antigen, as well as enhancing functionality using IL-12/15/18 cytokine pre-activation.We assessed single-cell NK cell responses stimulated by the tetravalent bispecific antibody AFM13 that binds CD30 on leukemia/lymphoma targets and CD16A on various types of NK cells using mass cytometry and cytotoxicity assays. The combination of AFM13 and IL-12/15/18 pre-activation of blood and cord-blood-derived NK cells was investigated in vitro and in vivo.We found heterogeneity within AFM13-directed conventional blood NK cell (cNK) responses, as well as consistent AFM13-directed polyfunctional activation of mature NK cells across donors. NK cell source also impacted the AFM13 response, with cNK cells from healthy donors exhibiting superior responses to those from Hodgkin lymphoma patients. IL-12/15/18-induced memory-like NK cells from peripheral blood exhibited enhanced killing of CD30+ lymphoma targets directed by AFM13, compared to cNK cells. Cord-blood NK cells pre-activated with IL-12/15/18 and ex vivo expanded with K562-based feeders also exhibited enhanced killing with AFM13 stimulation via upregulation of signaling pathways related to NK cell effector function. AFM13-NK complex cells exhibited enhanced responses to CD30+ lymphomas in vitro and in vivo.We identify AFM13 as a promising combination with cytokine-activated adult blood or cord blood NK cells to treat CD30+ hematologic malignancies, warranting clinical trials with these novel combinations.Copyright ©2021, American Association for Cancer Research.
[13]
Labrijn A F, Janmaat M L, Reichert J M, et al. Bispecific antibodies: a mechanistic review of the pipeline. Nature Reviews Drug Discovery, 2019, 18(8): 585-608.
The term bispecific antibody (bsAb) is used to describe a large family of molecules designed to recognize two different epitopes or antigens. BsAbs come in many formats, ranging from relatively small proteins, merely consisting of two linked antigen-binding fragments, to large immunoglobulin G (IgG)-like molecules with additional domains attached. An attractive bsAb feature is their potential for novel functionalities - that is, activities that do not exist in mixtures of the parental or reference antibodies. In these so-called obligate bsAbs, the physical linkage of the two binding specificities creates a dependency that can be temporal, with binding events occurring sequentially, or spatial, with binding events occurring simultaneously, such as in linking an effector to a target cell. To date, more than 20 different commercialized technology platforms are available for bsAb creation and development, 2 bsAbs are marketed and over 85 are in clinical development. Here, we review the current bsAb landscape from a mechanistic perspective, including a comprehensive overview of the pipeline.
[14]
Dennis M S, Zhang M, Meng Y G, et al. Albumin binding as a general strategy for improving the pharmacokinetics of proteins. Journal of Biological Chemistry, 2002, 277(38): 35035-35043.
Plasma protein binding can be an effective means of improving the pharmacokinetic properties of otherwise short lived molecules. Using peptide phage display, we identified a series of peptides having the core sequence DICLPRWGCLW that specifically bind serum albumin from multiple species with high affinity. These peptides bind to albumin with 1:1 stoichiometry at a site distinct from known small molecule binding sites. Using surface plasmon resonance, the dissociation equilibrium constant of peptide SA21 (Ac-RLIEDICLPRWGCLWEDD-NH(2)) was determined to be 266 +/- 8, 320 +/- 22, and 467 +/- 47 nm for rat, rabbit, and human albumin, respectively. SA21 has an unusually long half-life of 2.3 h when injected by intravenous bolus into rabbits. A related sequence, fused to the anti-tissue factor Fab of D3H44 (Presta, L., Sims, P., Meng, Y. G., Moran, P., Bullens, S., Bunting, S., Schoenfeld, J., Lowe, D., Lai, J., Rancatore, P., Iverson, M., Lim, A., Chisholm, V., Kelley, R. F., Riederer, M., and Kirchhofer, D. (2001) Thromb. Haemost. 85, 379-389), enabled the Fab to bind albumin with similar affinity to that of SA21 while retaining the ability of the Fab to bind tissue factor. This interaction with albumin resulted in reduced in vivo clearance of 25- and 58-fold in mice and rabbits, respectively, when compared with the wild-type D3H44 Fab. The half-life was extended 37-fold to 32.4 h in rabbits and 26-fold to 10.4 h in mice, achieving 25-43% of the albumin half-life in these animals. These half-lives exceed those of a Fab'(2) and are comparable with those seen for polyethylene glycol-conjugated Fab molecules, immunoadhesins, and albumin fusions, suggesting a novel and generic method for improving the pharmacokinetic properties of rapidly cleared proteins.
[15]
Bluemel C, Hausmann S, Fluhr P, et al. Epitope distance to the target cell membrane and antigen size determine the potency of T cell-mediated lysis by BiTE antibodies specific for a large melanoma surface antigen. Cancer Immunology, Immunotherapy, 2010, 59(8): 1197-1209.
[16]
Leong S R, Sukumaran S, Hristopoulos M, et al. An anti-CD3/anti-CLL-1 bispecific antibody for the treatment of acute myeloid leukemia. Blood, 2017, 129(5): 609-618.
Acute myeloid leukemia (AML) is a major unmet medical need. Most patients have poor long-term survival, and treatment has not significantly changed in 40 years. Recently, bispecific antibodies that redirect the cytotoxic activity of effector T cells by binding to CD3, the signaling component of the T-cell receptor, and a tumor target have shown clinical activity. Notably, blinatumomab is approved to treat relapsed/refractory acute lymphoid leukemia. Here we describe the design, discovery, pharmacologic activity, pharmacokinetics, and safety of a CD3 T cell-dependent bispecific (TDB) full-length human IgG1 therapeutic antibody targeting CLL-1 that could potentially be used in humans to treat AML. CLL-1 is prevalent in AML and, unlike other targets such as CD33 and CD123, is not expressed on hematopoietic stem cells providing potential hematopoietic recovery. We selected a high-affinity monkey cross-reactive anti-CLL-1 arm and tested several anti-CD3 arms that varied in affinity, and determined that the high-affinity CD3 arms were up to 100-fold more potent in vitro. However, in mouse models, the efficacy differences were less pronounced, probably because of prolonged exposure to TDB found with lower-affinity CD3 TDBs. In monkeys, assessment of safety and target cell depletion by the high- and low-affinity TDBs revealed that only the low-affinity CD3/CLL1 TDB was well tolerated and able to deplete target cells. Our data suggest that an appropriately engineered CLL-1 TDB could be effective in the treatment of AML.© 2017 by The American Society of Hematology.
[17]
Betts A, van der Graaf P H. Mechanistic quantitative pharmacology strategies for the early clinical development of bispecific antibodies in oncology. Clinical Pharmacology & Therapeutics, 2020, 108(3): 528-541.
[18]
Bacac M, Fauti T, Sam J, et al. A novel carcinoembryonic antigen T-cell bispecific antibody (CEA TCB) for the treatment of solid tumors. Clin Cancer Res, 2016; 22(13):3286-3297.
CEA TCB is a novel IgG-based T-cell bispecific (TCB) antibody for the treatment of CEA-expressing solid tumors currently in phase I clinical trials (NCT02324257). Its format incorporates bivalent binding to CEA, a head-to-tail fusion of CEA- and CD3e-binding Fab domains and an engineered Fc region with completely abolished binding to FcγRs and C1q. The study provides novel mechanistic insights into the activity and mode of action of CEA TCB.CEA TCB activity was characterized on 110 cell lines in vitro and in xenograft tumor models in vivo using NOG mice engrafted with human peripheral blood mononuclear cells.Simultaneous binding of CEA TCB to tumor and T cells leads to formation of immunologic synapses, T-cell activation, secretion of cytotoxic granules, and tumor cell lysis. CEA TCB activity strongly correlates with CEA expression, with higher potency observed in highly CEA-expressing tumor cells and a threshold of approximately 10,000 CEA-binding sites/cell, which allows distinguishing between high- and low-CEA-expressing tumor and primary epithelial cells, respectively. Genetic factors do not affect CEA TCB activity confirming that CEA expression level is the strongest predictor of CEA TCB activity. In vivo, CEA TCB induces regression of CEA-expressing xenograft tumors with variable amounts of immune cell infiltrate, leads to increased frequency of activated T cells, and converts PD-L1 negative into PD-L1-positive tumors.CEA TCB is a novel generation TCB displaying potent antitumor activity; it is efficacious in poorly infiltrated tumors where it increases T-cell infiltration and generates a highly inflamed tumor microenvironment. Clin Cancer Res; 22(13); 3286-97. ©2016 AACR.©2016 American Association for Cancer Research.
[19]
Wu X F, Sereno A J, Huang F, et al. Fab-based bispecific antibody formats with robust biophysical properties and biological activity. mAbs, 2015, 7(3): 470-482.
A myriad of innovative bispecific antibody (BsAb) platforms have been reported. Most require significant protein engineering to be viable from a development and manufacturing perspective. Single-chain variable fragments (scFvs) and diabodies that consist only of antibody variable domains have been used as building blocks for making BsAbs for decades. The drawback with Fv-only moieties is that they lack the native-like interactions with CH1/CL domains that make antibody Fab regions stable and soluble. Here, we utilize a redesigned Fab interface to explore 2 novel Fab-based BsAbs platforms. The redesigned Fab interface designs limit heavy and light chain mixing when 2 Fabs are co-expressed simultaneously, thus allowing the use of 2 different Fabs within a BsAb construct without the requirement of one or more scFvs. We describe the stability and activity of a HER2×HER2 IgG-Fab BsAb, and compare its biophysical and activity properties with those of an IgG-scFv that utilizes the variable domains of the same parental antibodies. We also generated an EGFR × CD3 tandem Fab protein with a similar format to a tandem scFv (otherwise known as a bispecific T cell engager or BiTE). We show that the Fab-based BsAbs have superior biophysical properties compared to the scFv-based BsAbs. Additionally, the Fab-based BsAbs do not simply recapitulate the activity of their scFv counterparts, but are shown to possess unique biological activity.
[20]
Luo Y D, Ye S, Li X K, et al. Emerging structure-function paradigm of endocrine FGFs in metabolic diseases. Trends in Pharmacological Sciences, 2019, 40(2): 142-153.
Endocrine fibroblast growth factors (eFGFs) control pathways that are crucial for maintaining metabolic homeostasis of lipids, glucose, energy, bile acids, and minerals. Unlike the heparin-binding paracrine FGFs, eFGFs require a unique Klotho family protein to form a productive triad complex, but the structural and mechanistical details of this complex have remained obscure since the beginning of the eFGF field. However, recent breakthroughs in resolving the 3D structures of eFGF signaling complexes have now unveiled the atomic details of multivalent interactions among eFGF, FGFR, and Klotho. We provide here a timely review on the architecture and the structure-function relationships of these complexes, and highlight how the structural knowledge opens a new door to structure-based drug design against a repertoire of eFGF-associated metabolic diseases.Copyright © 2018 Elsevier Ltd. All rights reserved.
[21]
Wu L J, Huang Y W, Sienkiewicz J, et al. Bispecific BCMA-CD3 antibodies block multiple myeloma tumor growth. Cancers, 2022, 14(10): 2518.
BCMA antigen is overexpressed in multiple myeloma cells and has been shown to be a promising target for novel cellular and antibody therapeutics. The humanized BCMA (clone 4C8A) antibody that effectively targeted multiple myeloma in a CAR (chimeric antigen receptor) format was used for designing several formats of bispecific BCMA-CD3 antibodies. Several different designs of univalent and bivalent humanized BCMA-CD3 CrossMAB and BCMA-FAB-CD3 ScFv-Fc antibodies were tested for binding with BCMA-positive cells and T cells and for killing by real time cytotoxic activity and IFN-gamma secretion with CHO-BCMA target cells and with multiple myeloma MM1S and H929 cell lines. All BCMA-CD3 antibodies demonstrated specific binding by FACS to CHO-BCMA, multiple myeloma cells, and to T cells with affinity Kd in the nM range. All antibodies with T cells specifically killed CHO-BCMA and multiple myeloma cells in a dose-dependent manner. The BCMA-CD3 antibodies with T cells secreted IFN-gamma with EC50 in the nM range. In addition, three BCMA bispecific antibodies had high in vivo efficacy using an MM1S xenograft NSG mouse model. The data demonstrate the high efficacy of novel hBCMA-CD3 antibodies with multiple myeloma cells and provide a basis for future pre-clinical and clinical development.
[22]
Seung E, Xing Z, Wu L, et al. A trispecific antibody targeting HER2 and T cells inhibits breast cancer growth via CD4 cells. Nature, 2022, 603(7900): 328-334.
[23]
Wu L, Seung E, Xu L, et al. Trispecific antibodies enhance the therapeutic efficacy of tumor-directed T cells through T cell receptor co-stimulation. Nature Cancer, 2019, 1(1): 86-98.
[24]
Damato B E, Dukes J, Goodall H, et al. Tebentafusp: T cell redirection for the treatment of metastatic uveal melanoma. Cancers, 2019, 11(7): 971.
Metastatic disease from uveal melanoma occurs in almost 50% of patients suffering from this ocular tumour, with median survival from development of symptoms being around 1 year. In contrast to cutaneous melanoma, kinase inhibitors and immune checkpoint inhibitors are usually ineffective in patients with metastatic uveal melanoma. Tebentafusp is a novel form of immunotherapy based on the immune-mobilising monoclonal T cell receptor against cancer (ImmTAC) platform, which comprises a soluble T cell receptor that is fused to an anti-CD3 single-chain variable fragment. The T cell receptor domain of tebentafusp targets cells present a human leukocyte antigen-A*02:01 complexed with a peptide derived from the melanoma-associated antigen gp100, which is expressed strongly by melanoma cells, weakly by normal melanocytes and minimally by other tissues. The anti-CD3 domain recruits CD3+ T cells (and, indirectly, other immune cells), redirecting these to the melanoma cells. The most common adverse events with tebentafusp are manageable and usually transient. Early survival data in patients with metastatic uveal melanoma are promising when considered alongside historical data. Based on these encouraging results, a randomised study comparing tebentafusp to investigator’s choice of therapy in metastatic uveal melanoma is ongoing.
[25]
Boustany L M, LaPorte S L, Wong L, et al. A Probody T cell-engaging bispecific antibody targeting EGFR and CD 3 inhibits colon cancer growth with limited toxicity. Cancer Res, 2022, 82(22):4288-4298.
T cell-engaging bispecific antibodies (TCB) are highly potent therapeutics that can recruit and activate cytotoxic T cells to stimulate an antitumor immune response. However, the development of TCBs against solid tumors has been limited by significant on-target toxicity to normal tissues. Probody therapeutics have been developed as a novel class of recombinant, protease-activated antibody prodrugs that are "masked" to reduce antigen binding in healthy tissues but can become conditionally unmasked by proteases that are preferentially active in the tumor microenvironment (TME). Here, we describe the preclinical efficacy and safety of CI107, a Probody TCB targeting EGFR and CD3. In vitro, the protease-activated, unmasked CI107 effectively bound EGFR and CD3 expressed on the surface of cells and induced T-cell activation, cytokine release, and cytotoxicity toward tumor cells. In contrast, dually masked CI107 displayed a >500-fold reduction in antigen binding and >15,000-fold reduction in cytotoxic activity. In vivo, CI107 potently induced dose-dependent tumor regression of established colon cancer xenografts in mice engrafted with human peripheral blood mononuclear cells. Furthermore, the MTD of CI107 in cynomolgus monkeys was more than 60-fold higher than that of the unmasked TCB, and much lower levels of toxicity were observed in animals receiving CI107. Therefore, by localizing activity to the TME and thus limiting toxicity to normal tissues, this Probody TCB demonstrates the potential to expand clinical opportunities for TCBs as effective anticancer therapies for solid tumor indications.A conditionally active EGFR-CD3 T cell-engaging Probody therapeutic expands the safety window of bispecific antibodies while maintaining efficacy in preclinical solid tumor settings.©2022 The Authors; Published by the American Association for Cancer Research.
[26]
Maude S L, Teachey D T, Porter D L, et al. CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood, 2015, 125(26): 4017-4023.
Relapsed and refractory acute lymphoblastic leukemia (ALL) remains difficult to treat, with minimal improvement in outcomes seen in more than 2 decades despite advances in upfront therapy and improved survival for de novo ALL. Adoptive transfer of T cells engineered to express a chimeric antigen receptor (CAR) has emerged as a powerful targeted immunotherapy, showing striking responses in highly refractory populations. Complete remission (CR) rates as high as 90% have been reported in children and adults with relapsed and refractory ALL treated with CAR-modified T cells targeting the B-cell-specific antigen CD19. Distinct CAR designs across several studies have produced similar promising CR rates, an encouraging finding. Even more encouraging are durable remissions observed in some patients without additional therapy. Duration of remission and CAR-modified T-cell persistence require further study and more mature follow-up, but emerging data suggest these factors may distinguish CAR designs. Supraphysiologic T-cell proliferation, a hallmark of this therapy, contributes to both efficacy and the most notable toxicity, cytokine release syndrome (CRS), posing a unique challenge for toxicity management. This review will discuss the current landscape of CD19 CAR clinical trials, CRS pathophysiology and management, and remaining challenges. © 2015 by The American Society of Hematology.
[27]
Strohl W R, Naso M. Bispecific T-cell redirection versus chimeric antigen receptor (CAR)-T cells as approaches to kill cancer cells. Antibodies, 2019, 8(3): 41.
The concepts for T-cell redirecting bispecific antibodies (TRBAs) and chimeric antigen receptor (CAR)-T cells are both at least 30 years old but both platforms are just now coming into age. Two TRBAs and two CAR-T cell products have been approved by major regulatory agencies within the last ten years for the treatment of hematological cancers and an additional 53 TRBAs and 246 CAR cell constructs are in clinical trials today. Two major groups of TRBAs include small, short-half-life bispecific antibodies that include bispecific T-cell engagers (BiTE®s) which require continuous dosing and larger, mostly IgG-like bispecific antibodies with extended pharmacokinetics that can be dosed infrequently. Most CAR-T cells today are autologous, although significant strides are being made to develop off-the-shelf, allogeneic CAR-based products. CAR-Ts form a cytolytic synapse with target cells that is very different from the classical immune synapse both physically and mechanistically, whereas the TRBA-induced synapse is similar to the classic immune synapse. Both TRBAs and CAR-T cells are highly efficacious in clinical trials but both also present safety concerns, particularly with cytokine release syndrome and neurotoxicity. New formats and dosing paradigms for TRBAs and CAR-T cells are being developed in efforts to maximize efficacy and minimize toxicity, as well as to optimize use with both solid and hematologic tumors, both of which present significant challenges such as target heterogeneity and the immunosuppressive tumor microenvironment.
[28]
Li J, Piskol R, Ybarra R, et al. CD3 bispecific antibody-induced cytokine release is dispensable for cytotoxic T cell activity. Science Translational Medicine, 2019, 11(508): eaax8861.
Cytokine release induced by anti-HER2/CD3 is mediated by TNF-α and can be suppressed by blocking TNF-α, IL-6, or IL-1 without affecting efficacy.
[29]
Norelli M, Camisa B, Barbiera G, et al. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nature Medicine, 2018, 24(6): 739-748.
In the clinic, chimeric antigen receptor-modified T (CAR T) cell therapy is frequently associated with life-threatening cytokine-release syndrome (CRS) and neurotoxicity. Understanding the nature of these pathologies and developing treatments for them are hampered by the lack of appropriate animal models. Herein, we describe a mouse model recapitulating key features of CRS and neurotoxicity. In humanized mice with high leukemia burden, CAR T cell-mediated clearance of cancer triggered high fever and elevated IL-6 levels, which are hallmarks of CRS. Human monocytes were the major source of IL-1 and IL-6 during CRS. Accordingly, the syndrome was prevented by monocyte depletion or by blocking IL-6 receptor with tocilizumab. Nonetheless, tocilizumab failed to protect mice from delayed lethal neurotoxicity, characterized by meningeal inflammation. Instead, the IL-1 receptor antagonist anakinra abolished both CRS and neurotoxicity, resulting in substantially extended leukemia-free survival. These findings offer a therapeutic strategy to tackle neurotoxicity and open new avenues to safer CAR T cell therapies.
[30]
Iwata Y, Sasaki M, Harada A, et al. Daily ascending dosing in cynomolgus monkeys to mitigate cytokine release syndrome induced by ERY22, surrogate for T-cell redirecting bispecific antibody ERY974 for cancer immunotherapy. Toxicology and Applied Pharmacology, 2019, 379: 114657.
[31]
Deppisch N, Ruf P, Eissler N, et al. Efficacy and tolerability of a GD2-directed trifunctional bispecific antibody in a preclinical model: subcutaneous administration is superior to intravenous delivery. Molecular Cancer Therapeutics, 2015, 14(8): 1877-1883.
Trifunctional bispecific antibodies (trAb) are novel anticancer drugs that recruit and activate different types of immune effector cells at the targeted tumor. Thus, tumor cells are effectively eliminated and a long-lasting tumor-specific T-cell memory is induced. The trAb Ektomab is directed against human CD3 on T cells and the tumor-associated ganglioside GD2, which is an attractive target for immunotherapy of melanoma in humans. To optimize clinical applicability, we studied different application routes with respect to therapeutic efficacy and tolerability by using the surrogate trAb Surek (anti-GD2 × anti-murine CD3) and a murine melanoma engineered to express GD2. We show that subcutaneous injection of the trAb is superior to the intravenous delivery pathway, which is the standard application route for therapeutic antibodies. Despite lower plasma levels after subcutaneous administration, the same tumor-protective potential was observed in vivo compared with intravenous administration of Surek. However, subcutaneously delivered Surek showed better tolerability. This could be explained by a continuous release of the antibody leading to constant plasma levels and a delayed induction of proinflammatory cytokines. Importantly, the induction of counter-regulatory mechanisms was reduced after subcutaneous application. These findings are relevant for the clinical application of trifunctional bispecific antibodies and, possibly, also other immunoglobulin constructs. Mol Cancer Ther; 14(8); 1877-83. ©2015 AACR. ©2015 American Association for Cancer Research.
[32]
Trinklein N D, Pham D, Schellenberger U, et al. Efficient tumor killing and minimal cytokine release with novel T-cell agonist bispecific antibodies. mAbs, 2019, 11(4): 639-652.
T-cell-recruiting bispecific antibodies (T-BsAbs) have shown potent tumor killing activity in humans, but cytokine release-related toxicities have affected their clinical utility. The use of novel anti-CD3 binding domains with more favorable properties could aid in the creation of T-BsAbs with improved therapeutic windows. Using a sequence-based discovery platform, we identified new anti-CD3 antibodies from humanized rats that bind to multiple epitopes and elicit varying levels of T-cell activation. In T-BsAb format, 12 different anti-CD3 arms induce equivalent levels of tumor cell lysis by primary T-cells, but potency varies by a thousand-fold. Our lead CD3-targeting arm stimulates very low levels of cytokine release, but drives robust tumor antigen-specific killing in vitro and in a mouse xenograft model. This new CD3-targeting antibody underpins a next-generation T-BsAb platform in which potent cytotoxicity is uncoupled from high levels of cytokine release, which may lead to a wider therapeutic window in the clinic.
[33]
Staflin K, Zuch de Zafra C L, Schutt L K, et al. Target arm affinities determine preclinical efficacy and safety of anti-HER2/CD 3 bispecific antibody. JCI Insight, 2020, 5(7): e133757.
[34]
Vafa O, Trinklein N D. Perspective: designing T-cell engagers with better therapeutic windows. Frontiers in Oncology, 2020, 10: 446.
This perspective highlights the history and challenges of developing CD3-based bispecific T-cell engagers (TCEs) as cancer therapeutics as well as considerations and potential strategies for designing the next generation TCE molecules. The goal of this article is to raise awareness of natural T-cell biology and how to best harness the tumor cell killing capacity of cytotoxic T-cells with TCEs. In light of 30 years of concerted efforts to advance TCEs in early clinical development, many of the first-generation bispecific antibodies have exhibited lackluster safety, efficacy, and manufacturability profiles. As of January 2020, blinatumomab remains the only approved TCE. Many of the current set-backs in early clinical trials implicate the high-affinity CD3 binding domains employed and the respective bispecific platforms as potential culprits. The underlying conviction of the authors is that by taking corrective measures, TCEs can transform cancer therapy. Through openness, transparency, and much needed feedback from ongoing clinical studies, the field can continuously improve the design and effectiveness of next generation T-cell redirecting therapeutics.Copyright © 2020 Vafa and Trinklein.
[35]
Chen X Y, Kamperschroer C, Wong G, et al. A modeling framework to characterize cytokine release upon T-cell-engaging bispecific antibody treatment: methodology and opportunities. Clinical and Translational Science, 2019, 12(6): 600-608.
T-cell-engaging bispecific antibodies (T-BsAbs) are an important class of antibody therapeutics in immuno-oncology. T-BsAbs simultaneously bind to CD3 on T cells and a tumor-associated antigen on tumor cells, activate T cells, and redirect T cells' cytotoxicity against tumor cells. Cytokine release syndrome (CRS), a common dose-limiting adverse event for T-BsAbs, is associated with T-cell activation. A "priming" dose strategy (i.e., a lower initial dose followed by a higher maintenance dose) has been implemented in the clinic to mitigate CRS and to achieve efficacious doses with T-BsAbs. So far, the selection of the optimal priming dosing regimen is largely empirical. A "fit-for-purpose" semimechanistic pharmacokinetic/pharmacodynamic model was developed to characterize the cytokine release profiles upon T-BsAb treatment, including the priming effect observed with repeated dosing. This model can be utilized to simulate cytokine profiles following various dosing regimens and may assist the design of clinical dosing strategies for T-BsAbs programs.© 2019 Pfizer Inc. Clinical and Translational Science published by Wiley Periodicals Inc. on behalf of the American Society of Clinical Pharmacology & Therapeutics.
[36]
Budde L E, Sehn L H, Matasar M, et al. Safety and efficacy of mosunetuzumab, a bispecific antibody, in patients with relapsed or refractory follicular lymphoma: a single-arm, multicentre, phase 2 study. The Lancet Oncology, 2022, 23(8): 1055-1065.
[37]
Haber L, Olson K, Kelly M P, et al. Generation of T-cell-redirecting bispecific antibodies with differentiated profiles of cytokine release and biodistribution by CD 3 affinity tuning. Scientific Reports, 2021, 11: 14397.
T-cell-redirecting bispecific antibodies have emerged as a new class of therapeutic agents designed to simultaneously bind to T cells via CD3 and to tumor cells via tumor-cell-specific antigens (TSA), inducing T-cell-mediated killing of tumor cells. The promising preclinical and clinical efficacy of TSAxCD3 antibodies is often accompanied by toxicities such as cytokine release syndrome due to T-cell activation. How the efficacy and toxicity profile of the TSAxCD3 bispecific antibodies depends on the binding affinity to CD3 remains unclear. Here, we evaluate bispecific antibodies that were engineered to have a range of CD3 affinities, while retaining the same binding affinity for the selected tumor antigen. These agents were tested for their ability to kill tumor cells in vitro, and their biodistribution, serum half-life, and anti-tumor activity in vivo. Remarkably, by altering the binding affinity for CD3 alone, we can generate bispecific antibodies that maintain potent killing of TSA + tumor cells but display differential patterns of cytokine release, pharmacokinetics, and biodistribution. Therefore, tuning CD3 affinity is a promising method to improve the therapeutic index of T-cell-engaging bispecific antibodies.\n
[38]
Santich B H, Cheal S M, Ahmed M, et al. A self-assembling and disassembling (SADA) bispecific antibody (BsAb) platform for curative two-step pretargeted radioimmunotherapy. Clin Cancer Res, 2021, 27(2):532-541.
Many cancer treatments suffer from dose-limiting toxicities to vital organs due to poor therapeutic indices. To overcome these challenges we developed a novel multimerization platform that rapidly removes tumor-targeting proteins from the blood to substantially improve therapeutic index.The platform was designed as a fusion of a self-assembling and disassembling (SADA) domain to a tandem single-chain bispecific antibody (BsAb, anti-ganglioside GD2 × anti-DOTA). SADA-BsAbs were assessed with multiple tumor models using two-step pretargeted radioimmunotherapy (PRIT) to evaluate tumor uptake, dosimetry, and antitumor responses.SADA-BsAbs self-assembled into stable tetramers (220 kDa), but could also disassemble into dimers or monomers (55 kDa) that rapidly cleared via renal filtration and substantially reduced immunogenicity in mice. When used with rapidly clearing DOTA-caged PET isotopes, SADA-BsAbs demonstrated accurate tumor localization, dosimetry, and improved imaging contrast by PET/CT. When combined with therapeutic isotopes, two-step SADA-PRIT safely delivered massive doses of alpha-emitting (Ac, 1.48 MBq/kg) or beta-emitting (Lu, 6,660 MBq/kg) S-2-(4-aminobenzyl)-1,4,7,10-tetraazacyclododecane tetraacetic acid (DOTA) payloads to tumors, ablating them without any short-term or long-term toxicities to the bone marrow, kidneys, or liver.The SADA-BsAb platform safely delivered large doses of radioisotopes to tumors and demonstrated no toxicities to the bone marrow, kidneys, or liver. Because of its modularity, SADA-BsAbs can be easily adapted to most tumor antigens, tumor types, or drug delivery approaches to improve therapeutic index and maximize the delivered dose..©2020 American Association for Cancer Research.
[39]
Peng J Q, Zhu Q, Peng Z R, et al. Patients with positive HER-2 amplification advanced gastroesophageal junction cancer achieved complete response with combined chemotherapy of AK104/cadonilimab (PD-1/CTLA-4 bispecific): a case report. Front Immunol, 2022, 13:1049518.
[40]
Alinari L, Mahoney E, Patton J, et al. FTY720 increases CD74 expression and sensitizes mantle cell lymphoma cells to milatuzumab-mediated cell death. Blood, 2011, 118(26): 6893-6903.
Mantle cell lymphoma (MCL) is an aggressive B-cell malignancy with a short median survival despite multimodal therapy. FTY720, an immunosuppressive drug approved for the treatment of multiple sclerosis, promotes MCL cell death concurrent with down-modulation of phospho-Akt and cyclin D1 and subsequent cell-cycle arrest. However, the mechanism of FTY720-mediated MCL cell death remains to be fully clarified. In the present study, we show features of autophagy blockage by FTY720 treatment, including accumulation of autolysosomes and increased LC3-II and p62 levels. We also show that FTY720-induced cell death is mediated by lysosomal membrane permeabilization with subsequent translocation of lysosomal hydrolases to the cytosol. FTY720-mediated disruption of the autophagic-lysosomal pathway led to increased levels of CD74, a potential therapeutic target in MCL that is degraded in the lysosomal compartment. This finding provided rationale for examining combination therapy with FTY720 and milatuzumab, an anti-CD74 mAb. Treatment of MCL cell lines and primary tumor cells with FTY720 and milatuzumab resulted in statistically significant enhanced cell death, which was synergistic in blastic variant MCL cell lines. Significant in vivo therapeutic activity of combination treatment was also demonstrated in a preclinical, in vivo model of MCL. These findings support clinical evaluation of this combination in patients with MCL.
[41]
Harrington K, Freeman D J, Kelly B, et al. Optimizing oncolytic virotherapy in cancer treatment. Nature Reviews Drug Discovery, 2019, 18(9): 689-706.
In the wake of the success of modern immunotherapy, oncolytic viruses (OVs) are currently seen as a potential therapeutic option for patients with cancer who do not respond or fail to achieve durable responses following treatment with immune checkpoint inhibitors. OVs offer a multifaceted therapeutic platform because they preferentially replicate in tumour cells, can be engineered to express transgenes that augment their cytotoxic and immunostimulatory activities, and modulate the tumour microenvironment to optimize immune-mediated tumour eradication, both at locoregional and systemic sites of disease. Lysis of tumour cells releases tumour-specific antigens that trigger both the innate and adaptive immune systems. OVs also represent attractive combination partners with other systemically delivered agents by virtue of their highly favourable safety profiles. Rational combinations of OVs with different immune modifiers and/or antitumour agents, based on mechanisms of tumour resistance to immune-mediated attack, may benefit the large, currently underserved, population of patients who respond poorly to immune checkpoint inhibition.
[42]
Ma R, Li Z L, Chiocca E A, et al. The emerging field of oncolytic virus-based cancer immunotherapy. Trends in Cancer, 2023, 9(2): 122-139.
[43]
Yu F, Wang X B, Guo Z S, et al. T-cell engager-armed oncolytic vaccinia virus significantly enhances antitumor therapy. Molecular Therapy: the Journal of the American Society of Gene Therapy, 2014, 22(1): 102-111.
[44]
Wang Q M, Ma X Y, Wu H, et al. Oncolytic adenovirus with MUC16-BiTE shows enhanced antitumor immune response by reversing the tumor microenvironment in PDX model of ovarian cancer. OncoImmunology, 2022, 11(1): 2096362.
[45]
Belmontes B, Sawant D V, Zhong W, et al. Immunotherapy combinations overcome resistance to bispecific T cell engager treatment in T cell-cold solid tumors. Science Translational Medicine, 2021, 13(608): eabd1524.
Immune checkpoint blockade and 4-1BB agonism overcome primary resistance to bispecific T cell engager therapy in T cell–cold solid tumors.
[46]
Wunderlich M, Manning N, Sexton C, et al. PD-1 inhibition enhances blinatumomab response in a UCB/PDX model of relapsed pediatric B-cell acute lymphoblastic leukemia. Frontiers in Oncology, 2021, 11: 642466.
Immune therapies such as blinatumomab, CD19-directed bispecific CD3 T-cell Engager (BiTE), have resulted in significant improvements in outcomes for relapsed B-cell acute lymphoblastic leukemia (B-ALL). However, up to half of blinatumomab treated patients do not respond completely or relapse after therapy. As a result, there is a need to identify potential strategies to improve the efficacy of BiTE therapy. The anti-PD-1 antibody pembrolizumab has been shown to successfully activate T cells against a wide range of cancer types. Here, we tested the ability of umbilical cord blood (UCB) reconstituted mice to respond to blinatumomab therapy with or without concurrent pembrolizumab treatment. Humanized mice were engrafted with patient-derived xenograft (PDX) cells derived from pediatric and adolescent/young adult (AYA) B-ALL patients who had either failed to achieve remission with negative minimum residual disease (MRD negative) or experienced a relapse. Mock-treated humanized mice engrafted with PDX cells efficiently developed overt disease within 30 days of engraftment of B-ALL. However, single agent therapy with either blinatumomab or pembrolizumab reduced disease burden in engrafted mice, with some mice observed to be MRD negative after the 28-day treatment course. Combination therapy significantly improved the percentage of MRD negative mice and improved long-term survival and cure rates as compared to mice that were given blinatumomab alone. Importantly, no benefits were observed in treated mice that lacked human immune cell reconstitution. These results indicate that UCB-humanized NRGS mice develop activatable immune function, and UCB-humanized PDX leukemia models can be used in preclinical studies to evaluate specificity, efficacy, and cooperativity of immune therapies in B-ALL.

基金

* 博士后创新人才支持计划(BX20220189)
国家自然科学基金(32070940)

PDF(1469 KB)

2583

Accesses

0

Citation

Detail

段落导航
相关文章

/