Model-informed drug discovery and development

  • 文章类型: Journal Article
    定量系统药理学(QSP)方法被广泛应用于解决药物发现和开发中的各种基本问题。例如识别治疗剂的作用机制,患者分层,以及对疾病进展的机械理解。在这篇评论文章中,从2013年到2022年,我们使用对QSP出版物的调查显示了QSP建模应用的现状。我们还提供了一个使用盐皮质激素受体拮抗剂治疗的糖尿病肾病患者高钾血症风险评估的用例(MRA,肾素-血管紧张素-醛固酮系统抑制剂),作为后期临床发展的前瞻性模拟。用于生成糖尿病肾病虚拟患者的QSP模型用于定量评估非甾体MRA,Finerenone和apararenone,高钾血症的风险比类固醇MRA低,eplerenone.使用QSP模型的前瞻性模拟研究有助于在临床开发中优先考虑候选药物,并验证与风险-收益相关的基于机制的药理学概念。在进行大规模临床试验之前。
    The quantitative systems pharmacology (QSP) approach is widely applied to address various essential questions in drug discovery and development, such as identification of the mechanism of action of a therapeutic agent, patient stratification, and the mechanistic understanding of the progression of disease. In this review article, we show the current landscape of the application of QSP modeling using a survey of QSP publications over 10 years from 2013 to 2022. We also present a use case for the risk assessment of hyperkalemia in patients with diabetic nephropathy treated with mineralocorticoid receptor antagonists (MRAs, renin-angiotensin-aldosterone system inhibitors), as a prospective simulation of late clinical development. A QSP model for generating virtual patients with diabetic nephropathy was used to quantitatively assess that the nonsteroidal MRAs, finerenone and apararenone, have a lower risk of hyperkalemia than the steroidal MRA, eplerenone. Prospective simulation studies using a QSP model are useful to prioritize pharmaceutical candidates in clinical development and validate mechanism-based pharmacological concepts related to the risk-benefit, before conducting large-scale clinical trials.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    抗体的靶标在基于抗体的治疗和诊断的成功中起着重要作用。和疫苗开发。这种重要性集中在靶结合位点-表位上,其中表位选择作为设计思维的一部分,超越了使用全细胞或全蛋白免疫的传统抗原选择,可以对成功产生积极影响。通过纯化的重组蛋白生产和肽合成来展示有限/选定的表位,可以更容易地选择可以影响产生的抗体的功能的内在因素。这些因素中的许多源于表位的位置,其可以在细胞或分子水平上影响抗体与表位的可接近性。直接抑制靶抗原活性,尽管逃避突变,功能的保守性,甚至是非竞争性抑制位点。通过将预测抗原变化的新计算方法纳入模型知情的药物发现和开发中,优秀的疫苗和基于抗体的治疗或诊断可以很容易地设计,以减轻失败。有了详细的例子,这次审查突出了新的机会,因素,以及预测抗原变化的方法,以考虑明智的表位选择。
    The target of an antibody plays a significant role in the success of antibody-based therapeutics and diagnostics, and vaccine development. This importance is focused on the target binding site-epitope, where epitope selection as a part of design thinking beyond traditional antigen selection using whole cell or whole protein immunization can positively impact success. With purified recombinant protein production and peptide synthesis to display limited/selected epitopes, intrinsic factors that can affect the functioning of resulting antibodies can be more easily selected for. Many of these factors stem from the location of the epitope that can impact accessibility of the antibody to the epitope at a cellular or molecular level, direct inhibition of target antigen activity, conservation of function despite escape mutations, and even noncompetitive inhibition sites. By incorporating novel computational methods for predicting antigen changes to model-informed drug discovery and development, superior vaccines and antibody-based therapeutics or diagnostics can be easily designed to mitigate failures. With detailed examples, this review highlights the new opportunities, factors, and methods of predicting antigenic changes for consideration in sagacious epitope selection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    A fit-for-purpose structural and statistical model is the first major requirement in population pharmacometric model development. In this manuscript we discuss how this complex and computationally intensive task could benefit from supervised machine learning algorithms. We compared the classical pharmacometric approach with two machine learning methods, genetic algorithm and neural networks, in different scenarios based on simulated pharmacokinetic data. Genetic algorithm performance was assessed using a fitness function based on log-likelihood, whilst neural networks were trained using mean square error or binary cross-entropy loss. Machine learning provided a selection based only on statistical rules and achieved accurate selection. The minimization process of genetic algorithm was successful at allowing the algorithm to select plausible models. Neural network classification tasks achieved the most accurate results. Neural network regression tasks were less precise than neural network classification and genetic algorithm methods. The computational gain obtained by using machine learning was substantial, especially in the case of neural networks. We demonstrated that machine learning methods can greatly increase the efficiency of pharmacokinetic population model selection in case of large datasets or complex models requiring long run-times. Our results suggest that machine learning approaches can achieve a first fast selection of models which can be followed by more conventional pharmacometric approaches.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    在药物开发中,决策通常通过基于模型的方法来支持,例如基于生理的药代动力学(PBPK)。全球敏感性分析(GSA)正在用于模型知情推断的质量评估。然而,GSA中相关因素的纳入和解释已被证明是一个问题。在这里,我们开发并评估了一种用于处理GSA中相关因素的潜在变量方法。开发了一种方法,该方法通过三个独立因素的因果关系来描述两个模型输入之间的相关性:潜在变量和两个相关参数的唯一方差。潜在变量方法应用于一组代数模型和PBPK的案例。然后,将该方法与Sobol的GSA进行比较,假设没有相关性,Sobol的GSA与团体和Kucherenko方法。对于潜在变量方法,GSA用Sobol方法进行。通过使用潜在变量方法,有可能设计出一种独特而简单的敏感性指数解释,同时保持因素之间的相关性。比较方法要么考虑参数独立,将因变量分组为一个独特的因素,或者在敏感性指数的解释中存在困难。在要求GSA支持模型知情决策的情况下,潜在变量方法提供了一种实用的方法,在易于实施和可解释性方面,将GSA应用于具有不违反独立性假设的相关输入的模型。讨论了该方法的先决条件和局限性。
    In drug development decision-making is often supported through model-based methods, such as physiologically-based pharmacokinetics (PBPK). Global sensitivity analysis (GSA) is gaining use for quality assessment of model-informed inference. However, the inclusion and interpretation of correlated factors in GSA has proven an issue. Here we developed and evaluated a latent variable approach for dealing with correlated factors in GSA. An approach was developed that describes the correlation between two model inputs through the causal relationship of three independent factors: the latent variable and the unique variances of the two correlated parameters. The latent variable approach was applied to a set of algebraic models and a case from PBPK. Then, this method was compared to Sobol\'s GSA assuming no correlations, Sobol\'s GSA with groups and the Kucherenko approach. For the latent variable approach, GSA was performed with Sobol\'s method. By using the latent variable approach, it is possible to devise a unique and easy interpretation of the sensitivity indices while maintaining the correlation between the factors. Compared methods either consider the parameters independent, group the dependent variables into one unique factor or present difficulties in the interpretation of the sensitivity indices. In situations where GSA is called upon to support model-informed decision-making, the latent variable approach offers a practical method, in terms of ease of implementation and interpretability, for applying GSA to models with correlated inputs that does not violate the independence assumption. Prerequisites and limitations of the approach are discussed.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    近年来,我们利用多维生物和临床数据从实验环境到现实环境的能力呈指数级增长,改变了药物研究和开发。随着人工智能(AI)和机器学习(ML)应用的增加。以患者为中心的迭代正向和反向翻译是精准医学发现和发展的核心,从目标验证到药物治疗的优化。将高级分析集成到转化医学实践中,现在是充分利用各种大数据集中包含的信息的基本推动者,例如“组学”数据。如基因组的深层特征所示,转录组,蛋白质组,代谢组,微生物组,和曝光。在这篇评论中,我们概述了ML在药物发现和开发中的应用,与转化医学的三大战略支柱(目标,病人,剂量),并提供有关其改变学科科学和实践的潜力的观点。讨论了将ML方法整合到药物计量学学科中的机会,并将彻底改变基于模型的药物发现和开发的实践。最后,我们认为临床药理学的共同努力,生物信息学,和生物标记技术专家在跨职能团队设置中至关重要,以实现支持AI/ML的转化和精准医学的承诺。
    The exponential increase in our ability to harness multi-dimensional biological and clinical data from experimental to real-world settings has transformed pharmaceutical research and development in recent years, with increasing applications of artificial intelligence (AI) and machine learning (ML). Patient-centered iterative forward and reverse translation is at the heart of precision medicine discovery and development across the continuum from target validation to optimization of pharmacotherapy. Integration of advanced analytics into the practice of Translational Medicine is now a fundamental enabler to fully exploit information contained in diverse sources of big data sets such as \"omics\" data, as illustrated by deep characterizations of the genome, transcriptome, proteome, metabolome, microbiome, and exposome. In this commentary, we provide an overview of ML applications in drug discovery and development, aligned with the three strategic pillars of Translational Medicine (target, patient, dose) and offer perspectives on their potential to transform the science and practice of the discipline. Opportunities for integrating ML approaches into the discipline of Pharmacometrics are discussed and will revolutionize the practice of model-informed drug discovery and development. Finally, we posit that joint efforts of Clinical Pharmacology, Bioinformatics, and Biomarker Technology experts are vital in cross-functional team settings to realize the promise of AI/ML-enabled Translational and Precision Medicine.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    In 2005, Danhof and coauthors proposed a new biomarker classification in the context of the application of mechanism-based PKPD modeling. They defined the term \'biomarker\' as a measure that characterizes a drug-induced response, which is on the causal path between drug administration and clinical outcome. The biomarker classification identified seven categories that provide different insights into the kinetics of drug action, such as target site distribution, target engagement, or into the impact of the drug on physiology or disease. The original biomarker classification has been further modified into a translational biomarker scheme that is used as a communication tool for drug hunting teams to guide designing translational and early clinical development plans as part of an integrated model-informed drug discovery and development strategy. It promotes a dedicated discussion on the topic of the translational relevance of biomarkers and enables efficient identification of translational gaps and opportunities. Based on the elucidated PKPD characteristics exhibited by a novel drug and the kinetics of the investigated biomarker, prospective predictions can be made for the drug response under new conditions; translating from the preclinical arena to the clinical setting, from the healthy volunteer to the patient, or from an adult to an elderly or a child. These drug response predictions provide support to decisions on appropriate next steps in the development of the drug, while keeping clear line of sight on the potential to address unmet medical need. Moreover, this framework enables a transparent translational risk assessment for drug hunting projects, and as such can underpin decisions at program and portfolio level.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Modeling & simulation (M&S) methodologies are established quantitative tools, which have proven to be useful in supporting the research, development (R&D), regulatory approval, and marketing of novel therapeutics. Applications of M&S help design efficient studies and interpret their results in context of all available data and knowledge to enable effective decision-making during the R&D process. In this mini-review, we focus on two sets of modeling approaches: population-based models, which are well-established within the pharmaceutical industry today, and fall under the discipline of clinical pharmacometrics (PMX); and systems dynamics models, which encompass a range of models of (patho-)physiology amenable to pharmacological intervention, of signaling pathways in biology, and of substance distribution in the body (today known as physiologically-based pharmacokinetic models) - which today may be collectively referred to as quantitative systems pharmacology models (QSP). We next describe the convergence - or rather selected integration - of PMX and QSP approaches into \'middle-out\' drug-disease models, which retain selected mechanistic aspects, while remaining parsimonious, fit-for-purpose, and able to address variability and the testing of covariates. We further propose development opportunities for drug-disease systems models, to increase their utility and applicability throughout the preclinical and clinical spectrum of pharmaceutical R&D.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号