
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*
Challenges and Therapeutic Strategies for Bispecific T Cell-redirecting Antibodies in Tumor Treatment
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。
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.
T细胞重定向双特异性抗体 / 免疫治疗 / 肿瘤 {{custom_keyword}} /
Bispecific T cell-redirecting antibodies / Immunotherapy / Tumor {{custom_keyword}} /
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 |
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 |
[1] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[2] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[3] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[4] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[5] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[6] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[7] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[8] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[9] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[10] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[11] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[12] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[13] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[14] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[15] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[16] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[17] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[18] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[19] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[20] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[21] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[22] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[23] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[24] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[25] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[26] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[27] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[28] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[29] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[30] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[31] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[32] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[33] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[34] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[35] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[36] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[37] |
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
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[38] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[39] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[40] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[41] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[42] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[43] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[44] |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[45] |
Immune checkpoint blockade and 4-1BB agonism overcome primary resistance to bispecific T cell engager therapy in T cell–cold solid tumors.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
[46] |
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.
{{custom_citation.content}}
{{custom_citation.annotation}}
|
{{custom_ref.label}} |
{{custom_citation.content}}
{{custom_citation.annotation}}
|
/
〈 |
|
〉 |