首页> 美国卫生研究院文献>PLoS Computational Biology >Target Inhibition Networks: Predicting Selective Combinations of Druggable Targets to Block Cancer Survival Pathways
【2h】

Target Inhibition Networks: Predicting Selective Combinations of Druggable Targets to Block Cancer Survival Pathways

机译:目标抑制网络:预测可药物靶向的选择性组合,以阻断癌症的生存途径

代理获取
本网站仅为用户提供外文OA文献查询和代理获取服务,本网站没有原文。下单后我们将采用程序或人工为您竭诚获取高质量的原文,但由于OA文献来源多样且变更频繁,仍可能出现获取不到、文献不完整或与标题不符等情况,如果获取不到我们将提供退款服务。请知悉。
获取外文期刊封面目录资料

摘要

A recent trend in drug development is to identify drug combinations or multi-target agents that effectively modify multiple nodes of disease-associated networks. Such polypharmacological effects may reduce the risk of emerging drug resistance by means of attacking the disease networks through synergistic and synthetic lethal interactions. However, due to the exponentially increasing number of potential drug and target combinations, systematic approaches are needed for prioritizing the most potent multi-target alternatives on a global network level. We took a functional systems pharmacology approach toward the identification of selective target combinations for specific cancer cells by combining large-scale screening data on drug treatment efficacies and drug-target binding affinities. Our model-based prediction approach, named TIMMA, takes advantage of the polypharmacological effects of drugs and infers combinatorial drug efficacies through system-level target inhibition networks. Case studies in MCF-7 and MDA-MB-231 breast cancer and BxPC-3 pancreatic cancer cells demonstrated how the target inhibition modeling allows systematic exploration of functional interactions between drugs and their targets to maximally inhibit multiple survival pathways in a given cancer type. The TIMMA prediction results were experimentally validated by means of systematic siRNA-mediated silencing of the selected targets and their pairwise combinations, showing increased ability to identify not only such druggable kinase targets that are essential for cancer survival either individually or in combination, but also synergistic interactions indicative of non-additive drug efficacies. These system-level analyses were enabled by a novel model construction method utilizing maximization and minimization rules, as well as a model selection algorithm based on sequential forward floating search. Compared with an existing computational solution, TIMMA showed both enhanced prediction accuracies in cross validation as well as significant reduction in computation times. Such cost-effective computational-experimental design strategies have the potential to greatly speed-up the drug testing efforts by prioritizing those interventions and interactions warranting further study in individual cancer cases.
机译:药物开发中的最新趋势是确定可以有效修饰疾病相关网络的多个节点的药物组合或多靶标药物。这种多药理作用可通过协同和合成致死性相互作用攻击疾病网络,从而降低出现耐药性的风险。但是,由于潜在药物和靶标组合的数量呈指数增长,因此需要系统的方法来在全球网络级别上确定最有效的多靶点替代方案的优先级。通过结合有关药物治疗效率和药物-靶标结合亲和力的大规模筛选数据,我们采用了功能系统药理学方法来鉴定特定癌细胞的选择性靶标组合。我们名为TIMMA的基于模型的预测方法利用了药物的多药理作用,并通过系统级靶标抑制网络推断出组合药物的疗效。在MCF-7和MDA-MB-231乳腺癌以及BxPC-3胰腺癌细胞中的案例研究证明,靶标抑制模型如何系统性地探索药物与其靶标之间的功能相互作用,以最大程度地抑制给定癌症类型中的多种生存途径。 TIMMA预测结果已通过系统化的siRNA介导的选定靶标及其成对组合沉默进行了实验验证,显示出不仅能够单独或组合识别癌症生存必需的可药物激酶靶标的能力增强,而且还具有协同作用指示非累加药效的相互作用。这些系统级分析是通过利用最大化和最小化规则的新型模型构建方法以及基于顺序前向浮点搜索的模型选择算法实现的。与现有的计算解决方案相比,TIMMA既显示了交叉验证中增强的预测准确性,又显着减少了计算时间。这种具有成本效益的计算实验设计策略有可能通过优先考虑那些干预措施和相互作用,从而有必要在个别癌症病例中进行进一步研究,从而大大加快药物测试的速度。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
代理获取

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