Engineering of a trispecific tumor-targeted immunotherapy incorporating 4-1BB co-stimulation and PD-L1 blockade

被引:22
作者
Warmuth, Stefan [1 ]
Gunde, Tea [1 ]
Snell, Daniel [1 ]
Brock, Matthias [1 ]
Weinert, Christopher [1 ]
Simonin, Alexandre [1 ]
Hess, Christian [1 ]
Tietz, Julia [1 ]
Johansson, Maria [1 ]
Spiga, Fabio Mario [1 ]
Heiz, Robin [1 ]
Fluckiger, Naomi [1 ]
Wagen, Sandro [1 ]
Zeberer, Julia [1 ]
Diem, Dania [1 ]
Mahler, Dana [1 ]
Wickihalder, Belinda [1 ]
Muntwiler, Simone [1 ]
Chatterjee, Bithi [1 ]
Kuttner, Benjamin [1 ]
Bommer, Bettina [1 ]
Yaman, Yasemin [1 ]
Lichtlen, Peter [1 ]
Urech, David [1 ]
机构
[1] Numab Therapeut AG, Einsiedlerstr 34, CH-8820 Wadenswil, Switzerland
来源
ONCOIMMUNOLOGY | 2021年 / 10卷 / 01期
关键词
Immune checkpoint inhibitor; T-cell stimulation; cancer immunotherapy; trispecific antibodies; fusion protein; antibody fragment; xenograft model; non-human primate; CD8(+) T-CELLS; DOSE-ESCALATION; ANTIBODY; AGONIST; CANCER; ACTIVATION; EXPANSION; MOLECULE; ANTIGEN; SAFETY;
D O I
10.1080/2162402X.2021.2004661
中图分类号
R73 [肿瘤学];
学科分类号
100214 ;
摘要
Co-stimulatory 4-1BB receptors on tumor-infiltrating T cells are a compelling target for overcoming resistance to immune checkpoint inhibitors, but initial clinical studies of 4-1BB agonist mAbs were accompanied by liver toxicity. We sought to engineer a tri-specific antibody-based molecule that stimulates intratumoral 4-1BB and blocks PD-L1/PD-1 signaling without systemic toxicity and with clinically favorable pharmacokinetics. Recombinant fusion proteins were constructed using scMATCH3 technology and humanized antibody single-chain variable fragments against PD-L1, 4-1BB, and human serum albumin. Paratope affinities were optimized using single amino acid substitutions, leading to design of the drug candidate NM21-1480. Multiple in vitro experiments evaluated pharmacodynamic properties of NM21-1480, and syngeneic mouse tumor models assessed antitumor efficacy and safety of murine analogues. A GLP multiple-dose toxicology study evaluated its safety in non-human primates. NM21-1480 inhibited PD-L1/PD-1 signaling with a potency similar to avelumab, and it potently stimulated 4-1BB signaling only in the presence of PD-L1, while exhibiting an EC50 that was largely independent of PD-L1 density. NM21-1480 exhibited high efficacy for co-activation of pre-stimulated T cells and dendritic cells. In xenograft models in syngeneic mice, NM21-1480 induced tumor regression and tumor infiltration of T cells without causing systemic T-cell activation. A GLP toxicology study revealed no evidence of liver toxicity at doses up to 140 mg/kg, and pharmacokinetic studies in non-human primates suggested a plasma half-life in humans of up to 2 weeks. NM21-1480 has the potential to overcome checkpoint resistance by co-activating tumor-infiltrating lymphocytes without liver toxicity.
引用
收藏
页数:17
相关论文
共 50 条
  • [11] A novel combinatorial cancer immunotherapy poly-IC and blockade of the PD-1/PD-L1 pathway
    Nagato, Toshihiro
    Celis, Esteban
    ONCOIMMUNOLOGY, 2014, 3 (05)
  • [12] PD-1/PD-L1 blockade: Prospectives for immunotherapy in cancer and autoimmunity
    Hosseinzadeh, Ramin
    Feizisani, Fahimeh
    Shomali, Navid
    Abdelbasset, Walid Kamal
    Hemmatzadeh, Maryam
    Gholizadeh Navashenaq, Jamshid
    Jadidi-Niaragh, Farhad
    Bokov, Dmitry O.
    Janebifam, Morteza
    Mohammadi, Hamed
    IUBMB LIFE, 2021, 73 (11) : 1293 - 1306
  • [13] Tumor cells versus host immune cells: whose PD-L1 contributes to PD-1/PD-L1 blockade mediated cancer immunotherapy?
    Fei Tang
    Pan Zheng
    Cell & Bioscience, 8
  • [14] Inhibitory Effect of PD-1/PD-L1 and Blockade Immunotherapy in Leukemia
    Xing, Kai
    Zhou, Pan
    Li, Jiaojiao
    Liu, Miao
    Zhang, Wei Emma
    COMBINATORIAL CHEMISTRY & HIGH THROUGHPUT SCREENING, 2022, 25 (09) : 1399 - 1410
  • [15] Predictive biomarkers in PD-1/PD-L1 checkpoint blockade immunotherapy
    Meng, Xiangjiao
    Huang, Zhaoqin
    Teng, Feifei
    Xing, Ligang
    Yu, Jinming
    CANCER TREATMENT REVIEWS, 2015, 41 (10) : 868 - 876
  • [16] Tumor cells versus host immune cells: whose PD-L1 contributes to PD-1/PD-L1 blockade mediated cancer immunotherapy?
    Tang, Fei
    Zheng, Pan
    CELL AND BIOSCIENCE, 2018, 8
  • [17] Self-Assembled Peptide-Derived Proteolysis-Targeting Chimera (PROTAC) Nanoparticles for Tumor-Targeted and Durable PD-L1 Degradation in Cancer Immunotherapy
    Moon, Yujeong
    Cho, Hanhee
    Kim, Jinseong
    Song, Sukyung
    Park, Jung Yeon
    Min, Jin Young
    Han, Eun Hee
    Kim, Yongju
    Seong, Joon-Kyung
    Shim, Man Kyu
    Kim, Kwangmeyung
    ANGEWANDTE CHEMIE-INTERNATIONAL EDITION, 2025, 64 (05)
  • [18] PD-L1 antibody conjugated dihydrotanshinone I-loaded polymeric nanoparticle for targeted cancer immunotherapy combining PD-L1 blockade with immunogenic cell death
    Wang, Xue
    Jing, Ziqi
    Huang, Xiaobin
    Liu, Xiaoya
    Zhang, Yujie
    Wang, Zhijun
    Ma, Pengkai
    INTERNATIONAL JOURNAL OF PHARMACEUTICS, 2024, 667
  • [19] PD-1/PD-L1 Checkpoint Inhibitors in Tumor Immunotherapy
    Liu, Jinhua
    Chen, Zichao
    Li, Yaqun
    Zhao, Wenjie
    Wu, JiBiao
    Zhang, Zhen
    FRONTIERS IN PHARMACOLOGY, 2021, 12
  • [20] Polymeric PD-L1 blockade nanoparticles for cancer photothermal-immunotherapy
    Yu, Yunjian
    Li, Jie
    Song, Boyi
    Ma, Zhuang
    Zhang, Yufei
    Sun, Haonan
    Wei, Xiaosong
    Bai, Yayun
    Lu, Xueguang
    Zhang, Peng
    Zhang, Xinge
    BIOMATERIALS, 2022, 280