...
首页> 外文期刊>Journal of pharmacokinetics and pharmacodynamics >On translation of antibody drug conjugates efficacy from mouse experimental tumors to the clinic: A PK/PD approach
【24h】

On translation of antibody drug conjugates efficacy from mouse experimental tumors to the clinic: A PK/PD approach

机译:从小鼠实验肿瘤到临床的抗体药物偶联物功效的转换:PK / PD方法

获取原文
获取原文并翻译 | 示例

摘要

Objectives of the present investigation were: (1) to compare three literature reported tumor growth inhibition (TGI) pharmacodynamic (PD) models and propose an optimal new model that best describes the xenograft TGI data for antibody drug conjugates (ADC), (2) to translate efficacy of the ADC Trastuzumab-emtansine (T-DM1) from mice to patients using the optimized PD model, and (3) to apply the translational strategy to predict clinically efficacious concentrations of a novel in-house anti-5T4 ADC, A1mcMMAF. First, the performance of all four of the PD models (i.e. 3 literature reported + 1 proposed) was evaluated using TGI data of T-DM1 obtained from four different xenografts. Based on the estimates of the pharmacodynamic/pharmacokinetic (PK/PD) modeling, a secondary parameter representing the efficacy index of the drug was calculated, which is termed as the tumor static concentration (TSC). TSC values derived from all four of the models were compared with each other, and with literature reported values, to assess the performance of these models. Subsequently, using the optimized PK/PD model, PD parameters obtained from different cell lines, human PK, and the proposed translational strategy, clinically efficacious doses of T-DM1 were projected. The accuracy of projected efficacious dose range for T-DM1 was verified by comparison with the clinical doses. Aforementioned strategy was then applied to A1mcMMAF for projecting its efficacious concentrations in clinic. TSC values for A1mcMMAF, obtained by fitting TGI data from 4 different xenografts with the proposed PK/PD model, were estimated to range from 0.6 to 11.5 μg mL-1. Accordingly, the clinically efficacious doses for A1mcMMAF were projected retrospectively. All in all, the improved PD model and proposed translational strategy presented here suggest that appropriate correction for the clinical exposure and employing the TSC criterion can help translate mouse TGI data to predict first in human doses of ADCs.
机译:本研究的目的是:(1)比较三种文献报道的肿瘤生长抑制(TGI)药效学(PD)模型,并提出一个最佳的新模型,该模型能最好地描述抗体药物偶联物(ADC)的异种移植TGI数据,(2)使用优化的PD模型将ADC曲妥珠单抗-坦丹碱(T-DM1)的功效从小鼠转化为患者,以及(3)应用转化策略来预测新型内部抗5T4 ADC A1mcMMAF的临床有效浓度。首先,使用从四种不同异种移植物中获得的T-DM1的TGI数据评估了所有四种PD模型的性能(即,报告了3篇文献,提出了1篇)。基于药效学/药代动力学(PK / PD)模型的估计,计算出代表药物功效指数的次要参数,称为肿瘤静态浓度(TSC)。将来自所有四个模型的TSC值相互比较,并与文献报道的值进行比较,以评估这些模型的性能。随后,使用优化的PK / PD模型,从不同细胞系,人PK获得的PD参数以及拟议的翻译策略,预测了T-DM1的临床有效剂量。通过与临床剂量比较,验证了T-DM1预计有效剂量范围的准确性。然后将上述策略应用于A1mcMMAF,以预测其在临床上的有效浓度。通过使用拟议的PK / PD模型拟合来自4种不同异种移植物的TGI数据获得的A1mcMMAF的TSC值估计为0.6至11.5μgmL-1。因此,回顾性地预测了A1mcMMAF的临床有效剂量。总而言之,此处介绍的改进的PD模型和拟议的翻译策略表明,针对临床暴露情况进行适当的校正并采用TSC标准可以帮助翻译小鼠TGI数据,从而首先预测人类ADC的剂量。

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