当前位置: X-MOL 学术Mol. Cancer Ther. › 论文详情
Our official English website, www.x-mol.net, welcomes your feedback! (Note: you will need to create a separate account there.)
Pharmacological inhibitor of DNA-PK, M3814, potentiates radiotherapy and regresses human tumors in mouse models
Molecular Cancer Therapeutics ( IF 5.7 ) Pub Date : 2020-03-27 , DOI: 10.1158/1535-7163.mct-19-0734
Frank T Zenke 1 , Astrid Zimmermann 1 , Christian Sirrenberg 1 , Heike Dahmen 1 , Vladimir Kirkin 2 , Ulrich Pehl 3 , Thomas Grombacher 4 , Claudia Wilm 1 , Thomas Fuchss 3 , Christiane Amendt 1 , Lyubomir T Vassilev 5 , Andree Blaukat 1
Affiliation  

Physical and chemical DNA-damaging agents are used widely in the treatment of cancer. Double-strand break (DSB) lesions in DNA are the most deleterious form of damage and, if left unrepaired, can effectively kill cancer cells. DNA-dependent protein kinase (DNA-PK) is a critical component of nonhomologous end joining (NHEJ), one of the two major pathways for DSB repair. Although DNA-PK has been considered an attractive target for cancer therapy, the development of pharmacologic DNA-PK inhibitors for clinical use has been lagging. Here, we report the discovery and characterization of a potent, selective, and orally bioavailable DNA-PK inhibitor, M3814 (peposertib), and provide in vivo proof of principle for DNA-PK inhibition as a novel approach to combination radiotherapy. M3814 potently inhibits DNA-PK catalytic activity and sensitizes multiple cancer cell lines to ionizing radiation (IR) and DSB-inducing agents. Inhibition of DNA-PK autophosphorylation in cancer cells or xenograft tumors led to an increased number of persistent DSBs. Oral administration of M3814 to two xenograft models of human cancer, using a clinically established 6-week fractionated radiation schedule, strongly potentiated the antitumor activity of IR and led to complete tumor regression at nontoxic doses. Our results strongly support DNA-PK inhibition as a novel approach for the combination radiotherapy of cancer. M3814 is currently under investigation in combination with radiotherapy in clinical trials.

中文翻译:

DNA-PK 的药理学抑制剂 M3814,在小鼠模型中增强放射治疗并使人类肿瘤消退

物理和化学 DNA 损伤剂广泛用于治疗癌症。DNA 中的双链断裂 (DSB) 损伤是最有害的损伤形式,如果不加以修复,可以有效杀死癌细胞。DNA 依赖性蛋白激酶 (DNA-PK) 是非同源末端连接 (NHEJ) 的关键组成部分,是 DSB 修复的两个主要途径之一。尽管 DNA-PK 被认为是癌症治疗的一个有吸引力的靶点,但用于临床应用的药理学 DNA-PK 抑制剂的开发一直滞后。在这里,我们报告了一种有效、选择性和口服生物可利用的 DNA-PK 抑制剂 M3814 (peposertib) 的发现和表征,并为 DNA-PK 抑制作为一种新的联合放疗方法提供了体内原理证明。M3814 有效抑制 DNA-PK 催化活性并使多种癌细胞系对电离辐射 (IR) 和 DSB 诱导剂敏感。癌细胞或异种移植肿瘤中 DNA-PK 自磷酸化的抑制导致持续性 DSB 数量增加。使用临床确立的 6 周分次放疗计划,将 M3814 口服给药至两种人类癌症异种移植模型,可强烈增强 IR 的抗肿瘤活性,并在无毒剂量下使肿瘤完全消退。我们的结果强烈支持 DNA-PK 抑制作为癌症联合放疗的新方法。M3814 目前正在临床试验中结合放疗进行研究。癌细胞或异种移植肿瘤中 DNA-PK 自磷酸化的抑制导致持续性 DSB 数量增加。使用临床确立的 6 周分次放疗计划,将 M3814 口服给药至两种人类癌症异种移植模型,可强烈增强 IR 的抗肿瘤活性,并在无毒剂量下使肿瘤完全消退。我们的结果强烈支持 DNA-PK 抑制作为癌症联合放疗的新方法。M3814 目前正在临床试验中结合放疗进行研究。癌细胞或异种移植肿瘤中 DNA-PK 自磷酸化的抑制导致持续性 DSB 数量增加。使用临床确立的 6 周分次放疗计划,将 M3814 口服给药至两种人类癌症异种移植模型,可强烈增强 IR 的抗肿瘤活性,并在无毒剂量下使肿瘤完全消退。我们的结果强烈支持 DNA-PK 抑制作为癌症联合放疗的新方法。M3814 目前正在临床试验中结合放疗进行研究。强烈增强 IR 的抗肿瘤活性,并在无毒剂量下导致肿瘤完全消退。我们的结果强烈支持 DNA-PK 抑制作为癌症联合放疗的新方法。M3814 目前正在临床试验中结合放疗进行研究。强烈增强 IR 的抗肿瘤活性,并在无毒剂量下导致肿瘤完全消退。我们的结果强烈支持 DNA-PK 抑制作为癌症联合放疗的新方法。M3814 目前正在临床试验中结合放疗进行研究。
更新日期:2020-03-27
down
wechat
bug