首页> 外文会议>American Society for Mass Spectrometry Conference on Mass Spectrometry and Allied Topics >Identification of Pyroglutamate and O-linked Glycosylation on Anti-IL17A Peptide Anti-IL22 Antibody Bispecific Genetic Fusion
【24h】

Identification of Pyroglutamate and O-linked Glycosylation on Anti-IL17A Peptide Anti-IL22 Antibody Bispecific Genetic Fusion

机译:抗IL17A肽抗IL22抗体双特异性遗传融合鉴定吡葡酸酯和O型糖基化的鉴定

获取原文

摘要

In this study, we have generated bispecific therapeutic fusion proteins in mammalian cells by combining a peptide and an antibody into a single open reading frame. A neutralizing peptide directed against interleukin-17A (IL17A) was genetically fused to the N-termini of an anti-IL22 antibody, through either the light chain, heavy chain, or both chains (Fig 1.). While the resulting fusion proteins bound and inhibited IL22 with the same affinity and potency as the unmodified anti-IL22 antibody, the peptide modality in the fusion scaffold was not active in the cell-based assay. In-Vitro serum stability analysis followed by immunoprecipitation and mass spectroscopic identification revealed instability in the N-terminal peptide portion of the molecule (Fig.2). When a glutamine residue was introduced at the N-terminus, which can be cyclized to form pyroglutamate in mammalian cells, the IL17A neutralization activity of the fusion protein was partially restored. Interestingly, the mass spectroscopic analysis of the purified fusion protein revealed an unexpected O-linked glycosylation modification at threonine 5 of the anti-IL17A peptide (Fig.3). Subsequent removal of this posttranslational modification by site-directed mutagenesis drastically enhanced the IL17A binding affinity and neutralization potency for the resulting fusion protein (Fig.4). These results provide direct experimental evidence that post-translational modifications during protein biosynthesis along secretory pathways play critical roles in determining the structure and function of therapeutic proteins produced by mammalian cells. The newly engineered peptideantibody genetic fusion is promising for therapeutically targeting multiple antigens in a single antibody-like molecule.
机译:在这项研究中,我们已经通过肽和抗体组合成单个开放阅读框产生在哺乳动物细胞中的双特异性的治疗性融合蛋白。针对白介素17A(IL17A)的中和肽基因融合到抗IL-22抗体的N-末端,通过任一轻链,重链,或两条链(图1)。而所得到的融合蛋白结合,并用相同的亲和力和效力与未修饰的抗IL22抗体抑制IL-22,在融合支架肽模态是不是在基于细胞的测定活性。体外血清稳定性分析及随后的免疫沉淀和质谱鉴定揭示了在分子(图2)的N-末端肽部分的不稳定性。当一个谷氨酰胺残基在N-末端,其可以被环化以形成在哺乳动物细胞中引入焦谷氨酸,融合蛋白的中和IL17A的活性部分恢复。有趣的是,纯化的融合蛋白的质谱分析在苏氨酸5的抗-IL-17A肽(图3)的显示意想不到的O-连接的糖基化修饰。通过位点定向诱变随后除去该翻译后修饰的飞跃性地提高的IL17A结合亲和力和中和效力为得到的融合蛋白(图4)。这些结果提供了直接的实验证据表明,沿分泌途径蛋白的生物合成过程中的翻译后修饰在确定由哺乳动物细胞产生的治疗性蛋白质的结构和功能中发挥关键作用。在新改造的peptideantibody遗传融合是有希望用于在单个抗体样分子治疗靶向多种抗原。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
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