...
首页> 外文期刊>Journal for ImmunoTherapy of Cancer >P09.14?Blocking counterregulation of unfolded protein response by targeted protein synthesis inhibition produces highly synergistic cell death in several cancer entities
【24h】

P09.14?Blocking counterregulation of unfolded protein response by targeted protein synthesis inhibition produces highly synergistic cell death in several cancer entities

机译:p09.14?通过靶向蛋白质合成抑制阻断展开蛋白质反应的反应在几个癌症实体中产生高度协同的细胞死亡

获取原文

摘要

Background Because tumor cells have high proliferation rates the demand for energy on the one hand and proteins on the other hand is high. In line, protein folding machinery of the ER is heavily used. 2-Deoxyglucose (2-DG) not only blocks energy synthesis by inhibiting glycolysis but also blocks synthesis of mannosyl leading to impaired N-linked glycosylation, accumulation of misfolded proteins, and increased unfolded protein response (UPR). However, due to compensatory events, UPR-induced apoptosis is hampered. Therefore, we combined 2-DG with targeted protein synthesis inhibition by immunotoxins, consisting of an antibody and pseudomonas exotoxin, to enhance UPR mediated cell death. Materials and Methods Established cell lines and patient-derived B-ALL samples were treated in vitro with various protein synthesis inhibitors and UPR-inducers. Drug synergy was determined mathematically as fold-increase over additivity. Biochemical studies were performed using western blots. In vivo enhancement was tested using systemic xenograft models. Results The combination of Moxetumomab and 2-DG achieved a two to nine-fold synergy in vitro . Synergy was abrogated by the addition of Mannose suggesting UPR as cause of synergistic cell death. Similarly, Moxetumomab enhanced UPR-inducers Bortezomib and tunicamycin and protein synthesis inhibition by cycloheximide and puromycin enhanced 2-DG suggesting a conserved mechanism. Using HB21, an immunotoxin targeting human transferrin-receptor, breast cancer, hepatocellular carcinoma, and glioblastoma were sensitized to 2-DG induced cell death. Biochemically, 2-DG increased XBP-1-cleavage, expression of pro-apoptotic CHOP and of anti-apoptotic BIP. Moxetumomab, however, blocked the upregulation of BIP while maintaining CHOP correlating with synergistic increase in PARP-cleavage and apoptosis. In two systemic mouse models, bone marrow (BM) lymphoma infiltration was not reduced by 2-DG or tunicamycin alone but was reduced after treatment with Moxetumomab alone by 5-fold in the JeKo-1 and by 16-fold in the Ramos model, respectively. The combination of Moxetumomab and 2-DG achieved a three-fold synergy in the JeKo-1 model and achieved MRD-negative BM status in the Ramos model. Against patient-derived B-ALL of the Burkitt’s type, 2-DG and Moxetumomab were up to 5-fold more active in vitro and up to 7-fold more active in mouse xenografts in vivo . Conclusions Cell death after persisting unfolded protein response is synergistically enhanced by tumor-cell specific inhibition of protein synthesis against four distinct tumor entities at physiologically achievable concentrations. Our approach of immunotoxin-induced targeted protein synthesis inhibition opens a novel, so far undescribed therapeutic window which may warrant clinical evaluation. Disclosure Information F. Gsottberger: None. C. Meier: None. S. Petkovic: None. L. Mellenthin: None. M. Krumbholz: None. M. Metzler: None. A. Mackensen: None. F. Müller: None.
机译:背景,因为肿瘤细胞具有高增殖率,一方面的能量需求和另一方面的蛋白质高。在线,ER的蛋白质折叠机械被大量使用。 2-脱氧葡萄糖(2-DG)不仅通过抑制糖酵解阻断能量合成,而且还阻断了甘露糖基的合成,导致N-连接的糖基化受损,误折叠蛋白质的积累,以及增加的展开蛋白质反应(UPR)。然而,由于补偿事件,upr诱导的细胞凋亡受到阻碍。因此,我们将2-DG与ImmunoToxins的靶向蛋白质合成抑制合并,由Exotoxin组成,以增强UPR介导的细胞死亡。材料和方法建立了细胞系和患者衍生的B-所有样品,体外用各种蛋白质合成抑制剂和UPR诱导剂处理。药物协同作用在数学上确定为增加的添加性。使用蛋白质印迹进行生化研究。使用全身异种移植模型测试体内增强。结果Moxetumomab和2-Dg的组合在体外实现了两到9倍的协同作用。随着协同细胞死亡的原因,通过添加曼诺斯的曼诺斯提出了协同作用。类似地,Moxetumomab增强UPR-inducers Bortezomib和Tunicamycin和蛋白质合成抑制通过环己酰亚胺和嘌呤霉素增强2-DG表明保守机制。使用HB21,靶向人转移素受体,乳腺癌,肝细胞癌和胶质母细胞瘤的免疫毒素致敏于2-DG诱导的细胞死亡。生物化学上,2-DG增加XBP-1 - 切割,促凋亡斩波和抗凋亡蛋白的表达。然而,Moxetumomab阻断了BIP的上调,同时保持切碎与PARP切割和细胞凋亡的协同增加相关。在两个全身鼠标模型中,骨髓(BM)淋巴瘤渗透单独不会减少2-DG或unicamycin,但在用麦克苏忒ob在Jeko-1中以5倍和Ramos模型中的16倍后处理,分别。 Moxetumomab和2-DG的组合在JEKO-1模型中实现了三倍的协同作用,并在Ramos模型中实现了MRD阴性BM状态。针对患者衍生的B- Burkitt的类型,2-DG和Moxetumomab在体外体外高达5倍,在体内小鼠异种移植物中最活跃高达7倍。结论在持久性展开蛋白质反应后的细胞死亡通过在生理学上可实现的浓度下对四种不同的肿瘤实体进行肿瘤细胞特异性抑制协同增强。我们的免疫毒素诱导的靶向蛋白质合成抑制方法开启了一种新颖的,到目前为止的未描述的治疗窗口,可能需要临床评估。披露信息F. Gsottberger:无。 C. Meier:没有。 S. Petkovic:没有。 L. Mellenthin:没有。 M. Krumbholz:没有。 M. Metzler:没有。 A. Mackensen:没有。 F.Müller:没有。

著录项

获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号