当前位置: X-MOL 学术Pharm. Res. › 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.)
Application of Pharmacokinetic-Pharmacodynamic Modeling to Inform Translation of In Vitro NaV1.7 Inhibition to In Vivo Pharmacological Response in Non-human Primate.
Pharmaceutical Research ( IF 3.7 ) Pub Date : 2020-09-04 , DOI: 10.1007/s11095-020-02914-9
Jeanine E Ballard 1 , Parul Pall 2 , Joshua Vardigan 2 , Fuqiang Zhao 3 , Marie A Holahan 3 , Richard Kraus 4 , Yuxing Li 4 , Darrell Henze 5 , Andrea Houghton 5 , Christopher S Burgey 6 , Christopher Gibson 1
Affiliation  

Purpose

This work describes a staged approach to the application of pharmacokinetic-pharmacodynamic (PK-PD) modeling in the voltage-gated sodium ion channel (NaV1.7) inhibitor drug discovery effort to address strategic questions regarding in vitro to in vivo translation of target modulation.

Methods

PK-PD analysis was applied to data from a functional magnetic resonance imaging (fMRI) technique to non-invasively measure treatment mediated inhibition of olfaction signaling in non-human primates (NHPs). Initial exposure-response was evaluated using single time point data pooled across 27 compounds to inform on in vitro to in vivo correlation (IVIVC). More robust effect compartment PK-PD modeling was conducted for a subset of 10 compounds with additional PD and PK data to characterize hysteresis.

Results

The pooled compound exposure-response facilitated an early exploration of IVIVC with a limited dataset for each individual compound, and it suggested a 2.4-fold in vitro to in vivo scaling factor for the NaV1.7 target. Accounting for hysteresis with an effect compartment PK-PD model as compounds advanced towards preclinical development provided a more robust determination of in vivo potency values, which resulted in a statistically significant positive IVIVC with a slope of 1.057 ± 0.210, R-squared of 0.7831, and p value of 0.006. Subsequent simulations with the PK-PD model informed the design of anti-nociception efficacy studies in NHPs.

Conclusions

A staged approach to PK-PD modeling and simulation enabled integration of in vitro NaV1.7 potency, plasma protein binding, and pharmacokinetics to describe the exposure-response profile and inform future study design as the NaV1.7 inhibitor effort progressed through drug discovery.


中文翻译:

药代动力学-药效学建模在通知非人类灵长类动物体内NaV1.7抑制体内药理反应的翻译中的应用。

目的

这项工作描述了一种分阶段的方式来的药代动力学-药效学(PK-PD)的施加在电压-门控钠离子通道(NaV1.7)抑制剂药物发现关于努力地址战略问题建模体外体内靶调制的翻译。

方法

PK-PD分析应用于功能磁共振成像(fMRI)技术的数据,以非侵入性方式测量治疗介导的非人类灵长类动物(NHP)嗅觉信号的抑制。使用汇总在27种化合物中的单个时间点数据来评估初始暴露反应,以告知体外体内的相关性(IVIVC)。对10种化合物的子集进行了更鲁棒的效应区PK-PD建模,并附加了PD和PK数据以表征磁滞现象。

结果

合并的化合物暴露-反应促进了对每种化合物的有限数据集的IVIVC的早期探索,并且提出了NaV1.7靶标的2.4倍的体外-体内比例因子。随着化合物向临床前发展的进展,使用效应区室PK-PD模型解决了滞后现象,从而提供了更强大的体内效价测定方法,从而得出具有统计学意义的正IVIVC阳性斜率,斜率为1.057±0.210,R平方为0.7831,和p为0.006的值。随后用PK-PD模型进行的模拟为NHP中抗伤害感受功效研究的设计提供了依据。

结论

通过分阶段进行PK-PD建模和模拟,可以整合体外NaV1.7效价,血浆蛋白结合和药代动力学,以描述暴露-反应曲线,并随着NaV1.7抑制剂努力通过药物发现而发展,为将来的研究设计提供依据。
更新日期:2020-09-04
down
wechat
bug