基于模型的早期药物开发:用定量清晰度导航复杂性

IF 1.8 4区 医学 Q3 PHARMACOLOGY & PHARMACY
Amalia M. Issa
{"title":"基于模型的早期药物开发:用定量清晰度导航复杂性","authors":"Amalia M. Issa","doi":"10.1002/cpdd.1607","DOIUrl":null,"url":null,"abstract":"<p>Model-informed drug development (MIDD) has evolved from a promising innovation to a regulatory imperative in drug development. Over the past year, the regulatory landscape shifted. The Food and Drug Administration (FDA) finalized guidance on oncology dose optimization under Project Optimus<span><sup>1, 2</sup></span> and institutionalized its MIDD Paired Meeting Program,<span><sup>3</sup></span> while ICH issued E11A for pediatric extrapolation<span><sup>4</sup></span> and released a draft guideline M15, developed under ICH auspices, on generalized MIDD principles.<span><sup>5</sup></span> These developments collectively transform early drug development: the goal is no longer just identifying a tolerated dose but quantitatively justifying optimal dosing for pivotal trials. By integrating pharmacokinetics (PK), pharmacodynamics (PD), systems pharmacology, and real-world data, MIDD reduces development risks, accelerates first-in-human studies, and enables rational dose selection. Its expanding influence on decision-making underscores its role not only as a technical methodology but as a paradigm shift in early-phase development strategy.</p><p>In the last decade, the utility of MIDD has expanded significantly, driven by advances in computational modeling and an increasingly favorable regulatory environment. The FDA and the European Medicines Agency (EMA) have articulated frameworks encouraging the use of MIDD in early development, with the FDA's <i>MIDD Pilot Program</i> serving as a cornerstone initiative fostering sponsor–regulator dialogue.<span><sup>3, 5-7</sup></span> In 2025, such efforts have matured, with MIDD now recognized as central to regulatory submissions in oncology, rare diseases, and immunology.</p><p>Recent literature emphasizes that MIDD enables the translation of preclinical data into clinically relevant predictions with unprecedented precision. For example, Ren et al<span><sup>8</sup></span> highlight how PK/PD modeling refines dose optimization strategies in discovery and development, allowing earlier identification of optimal therapeutic windows.<span><sup>3</sup></span> This has relevance in first-in-human trials, where dose escalation strategies can now be informed by quantitative predictions rather than empirical trial-and-error. This shift reframes early-phase development from “first safe dose” thinking to “first scientifically informative dose.”</p><p>1. <i>First-in-Human Dose Prediction</i>. Physiologically based pharmacokinetic (PBPK) modeling has revolutionized first-in-human dose selection, particularly for biologics and small molecules with complex metabolism. PBPK models incorporate variability in absorption, metabolism, and clearance across virtual populations, offering mechanistic insights that go beyond traditional allometric scaling.<span><sup>9</sup></span> In oncology and rare diseases, such models are increasingly used to support starting dose rationalization, minimizing both underdosing and toxicity risk.</p><p>2. <i>Adaptive Trial Designs</i>. MIDD facilitates adaptive trial designs in Phase I studies. By integrating Bayesian hierarchical models with real-time PK/PD data, investigators can adjust dose escalation schemes dynamically. This reduces trial duration and exposure of participants to subtherapeutic or unsafe doses. The FDA's recent endorsements of adaptive MIDD frameworks underscore their regulatory acceptance.<span><sup>1, 3, 5</sup></span></p><p>3. <i>Drug–Drug Interaction (DDI) Risk Mitigation</i>. PBPK models are now routinely leveraged to predict cytochrome-450 (CYP)-mediated and transporter-mediated DDIs early in development, reducing the need for extensive dedicated clinical DDI studies.<span><sup>10</sup></span> This integration saves time and resources while providing mechanistic justifications to regulators.</p><p>4. <i>Rare Disease and Precision Medicine</i>. For rare diseases, where patient numbers are inherently small, MIDD supports extrapolation from sparse datasets to inform early-phase dosing strategies. Chen et al (2025) note that MIDD also provides a path for evaluating pharmacological modulation of post-translational modifications in cancer, where biomarker-driven modeling is critical for dose selection.<span><sup>11</sup></span></p><p>The growth of artificial intelligence (AI) and machine learning has further augmented MIDD. These technologies allow for real-time updating of population PK models with electronic health record data and wearable-derived biomarkers, enabling <i>continuous model refinement</i> during early-phase trials.<span><sup>12</sup></span> Hybrid approaches that combine mechanistic PBPK with data-driven AI models are becoming the standard, improving predictive accuracy across diverse populations.</p><p>Another frontier is quantitative systems pharmacology (QSP). QSP integrates network biology, omics data, and mechanistic modeling to understand drug–disease interactions. Its application in Phase I studies is expanding, particularly in immuno-oncology and metabolic disorders, where dynamic feedback loops complicate dose–response predictions.<span><sup>13</sup></span></p><p>It is one thing to laud MIDD's conceptual elegance; it is another to show tangible returns. Analyses have shown considerable savings both in terms of time and costs. Importantly, these savings are not just monetary or temporal—they reshape strategic decision-making. MIDD facilitates early “No-Go” decisions, reallocation of resources, and heightened confidence in dosing strategy. Automation tools such as Automated Monitoring of Phase I; 2 (AMP) for Phase I data processing and Cardio Exposure–Response Modeling (CardioERM) for concentration–QT analysis further compressed timelines, turning weeks of report generation into minutes and saving hundreds of business days annually.<span><sup>14</sup></span></p><p>The regulatory environment in 2025 actively embraces MIDD, publishing detailed guidance on the use of PBPK, population PK, and QSP in early development.<span><sup>3, 5, 6, 15, 16</sup></span> Regulatory science is increasingly collaborative, with workshops and pre-IND consultations dedicated to discussing model credibility. Importantly, regulators now expect sponsors to demonstrate how modeling influenced dose selection and trial design.</p><p>Ethically, MIDD reduces unnecessary human exposure to unsafe doses, aligning with the principle of minimizing participant risk. In rare and pediatric diseases, MIDD provides an ethical pathway for extrapolating dosing regimens from adult or preclinical data, reducing the burden on vulnerable populations.<span><sup>17</sup></span></p><p>Despite progress, challenges persist. Model credibility and reproducibility remain concerns, particularly when integrating AI-driven predictions. Transparency in modeling assumptions and rigorous qualification frameworks are essential to maintain trust among regulators and clinicians.<span><sup>12</sup></span> Moreover, the heterogeneity of data sources from preclinical models to real-world datasets poses integration challenges that require standardization.</p><p>The evolution of MIDD in early development parallels the growing adoption of AI and machine learning-enabled modeling approaches. As real-world data and in silico methods become increasingly integrated into preclinical-to-clinical translation, a shift toward what is now termed MID<sup>3</sup> (model-informed drug discovery and development)<span><sup>21</sup></span> is becoming evident. At this convergence point, investment in modeling expertise, infrastructure, and regulatory–industry collaboration is essential to sustain its impact.</p><p>MIDD represents not merely a technical toolkit but a paradigm shift for early-phase innovation. By integrating quantitative predictions, mechanistic modeling, and adaptive learning, MIDD de-risks early clinical development and establishes a stronger scientific foundation for subsequent phases. Regulatory willingness to embrace these approaches creates an opportunity to embed modeling as a standard, rather than exceptional, practice in first-in-human drug development.</p><p>The author declares no conflicts of interest.</p><p>No funding was obtained for this work.</p>","PeriodicalId":10495,"journal":{"name":"Clinical Pharmacology in Drug Development","volume":"14 10","pages":"738-741"},"PeriodicalIF":1.8000,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://accp1.onlinelibrary.wiley.com/doi/epdf/10.1002/cpdd.1607","citationCount":"0","resultStr":"{\"title\":\"Model-Informed Drug Development in Early-Phase Development: Navigating Complexity With Quantitative Clarity\",\"authors\":\"Amalia M. Issa\",\"doi\":\"10.1002/cpdd.1607\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>Model-informed drug development (MIDD) has evolved from a promising innovation to a regulatory imperative in drug development. Over the past year, the regulatory landscape shifted. The Food and Drug Administration (FDA) finalized guidance on oncology dose optimization under Project Optimus<span><sup>1, 2</sup></span> and institutionalized its MIDD Paired Meeting Program,<span><sup>3</sup></span> while ICH issued E11A for pediatric extrapolation<span><sup>4</sup></span> and released a draft guideline M15, developed under ICH auspices, on generalized MIDD principles.<span><sup>5</sup></span> These developments collectively transform early drug development: the goal is no longer just identifying a tolerated dose but quantitatively justifying optimal dosing for pivotal trials. By integrating pharmacokinetics (PK), pharmacodynamics (PD), systems pharmacology, and real-world data, MIDD reduces development risks, accelerates first-in-human studies, and enables rational dose selection. Its expanding influence on decision-making underscores its role not only as a technical methodology but as a paradigm shift in early-phase development strategy.</p><p>In the last decade, the utility of MIDD has expanded significantly, driven by advances in computational modeling and an increasingly favorable regulatory environment. The FDA and the European Medicines Agency (EMA) have articulated frameworks encouraging the use of MIDD in early development, with the FDA's <i>MIDD Pilot Program</i> serving as a cornerstone initiative fostering sponsor–regulator dialogue.<span><sup>3, 5-7</sup></span> In 2025, such efforts have matured, with MIDD now recognized as central to regulatory submissions in oncology, rare diseases, and immunology.</p><p>Recent literature emphasizes that MIDD enables the translation of preclinical data into clinically relevant predictions with unprecedented precision. For example, Ren et al<span><sup>8</sup></span> highlight how PK/PD modeling refines dose optimization strategies in discovery and development, allowing earlier identification of optimal therapeutic windows.<span><sup>3</sup></span> This has relevance in first-in-human trials, where dose escalation strategies can now be informed by quantitative predictions rather than empirical trial-and-error. This shift reframes early-phase development from “first safe dose” thinking to “first scientifically informative dose.”</p><p>1. <i>First-in-Human Dose Prediction</i>. Physiologically based pharmacokinetic (PBPK) modeling has revolutionized first-in-human dose selection, particularly for biologics and small molecules with complex metabolism. PBPK models incorporate variability in absorption, metabolism, and clearance across virtual populations, offering mechanistic insights that go beyond traditional allometric scaling.<span><sup>9</sup></span> In oncology and rare diseases, such models are increasingly used to support starting dose rationalization, minimizing both underdosing and toxicity risk.</p><p>2. <i>Adaptive Trial Designs</i>. MIDD facilitates adaptive trial designs in Phase I studies. By integrating Bayesian hierarchical models with real-time PK/PD data, investigators can adjust dose escalation schemes dynamically. This reduces trial duration and exposure of participants to subtherapeutic or unsafe doses. The FDA's recent endorsements of adaptive MIDD frameworks underscore their regulatory acceptance.<span><sup>1, 3, 5</sup></span></p><p>3. <i>Drug–Drug Interaction (DDI) Risk Mitigation</i>. PBPK models are now routinely leveraged to predict cytochrome-450 (CYP)-mediated and transporter-mediated DDIs early in development, reducing the need for extensive dedicated clinical DDI studies.<span><sup>10</sup></span> This integration saves time and resources while providing mechanistic justifications to regulators.</p><p>4. <i>Rare Disease and Precision Medicine</i>. For rare diseases, where patient numbers are inherently small, MIDD supports extrapolation from sparse datasets to inform early-phase dosing strategies. Chen et al (2025) note that MIDD also provides a path for evaluating pharmacological modulation of post-translational modifications in cancer, where biomarker-driven modeling is critical for dose selection.<span><sup>11</sup></span></p><p>The growth of artificial intelligence (AI) and machine learning has further augmented MIDD. These technologies allow for real-time updating of population PK models with electronic health record data and wearable-derived biomarkers, enabling <i>continuous model refinement</i> during early-phase trials.<span><sup>12</sup></span> Hybrid approaches that combine mechanistic PBPK with data-driven AI models are becoming the standard, improving predictive accuracy across diverse populations.</p><p>Another frontier is quantitative systems pharmacology (QSP). QSP integrates network biology, omics data, and mechanistic modeling to understand drug–disease interactions. Its application in Phase I studies is expanding, particularly in immuno-oncology and metabolic disorders, where dynamic feedback loops complicate dose–response predictions.<span><sup>13</sup></span></p><p>It is one thing to laud MIDD's conceptual elegance; it is another to show tangible returns. Analyses have shown considerable savings both in terms of time and costs. Importantly, these savings are not just monetary or temporal—they reshape strategic decision-making. MIDD facilitates early “No-Go” decisions, reallocation of resources, and heightened confidence in dosing strategy. Automation tools such as Automated Monitoring of Phase I; 2 (AMP) for Phase I data processing and Cardio Exposure–Response Modeling (CardioERM) for concentration–QT analysis further compressed timelines, turning weeks of report generation into minutes and saving hundreds of business days annually.<span><sup>14</sup></span></p><p>The regulatory environment in 2025 actively embraces MIDD, publishing detailed guidance on the use of PBPK, population PK, and QSP in early development.<span><sup>3, 5, 6, 15, 16</sup></span> Regulatory science is increasingly collaborative, with workshops and pre-IND consultations dedicated to discussing model credibility. Importantly, regulators now expect sponsors to demonstrate how modeling influenced dose selection and trial design.</p><p>Ethically, MIDD reduces unnecessary human exposure to unsafe doses, aligning with the principle of minimizing participant risk. In rare and pediatric diseases, MIDD provides an ethical pathway for extrapolating dosing regimens from adult or preclinical data, reducing the burden on vulnerable populations.<span><sup>17</sup></span></p><p>Despite progress, challenges persist. Model credibility and reproducibility remain concerns, particularly when integrating AI-driven predictions. Transparency in modeling assumptions and rigorous qualification frameworks are essential to maintain trust among regulators and clinicians.<span><sup>12</sup></span> Moreover, the heterogeneity of data sources from preclinical models to real-world datasets poses integration challenges that require standardization.</p><p>The evolution of MIDD in early development parallels the growing adoption of AI and machine learning-enabled modeling approaches. As real-world data and in silico methods become increasingly integrated into preclinical-to-clinical translation, a shift toward what is now termed MID<sup>3</sup> (model-informed drug discovery and development)<span><sup>21</sup></span> is becoming evident. At this convergence point, investment in modeling expertise, infrastructure, and regulatory–industry collaboration is essential to sustain its impact.</p><p>MIDD represents not merely a technical toolkit but a paradigm shift for early-phase innovation. By integrating quantitative predictions, mechanistic modeling, and adaptive learning, MIDD de-risks early clinical development and establishes a stronger scientific foundation for subsequent phases. Regulatory willingness to embrace these approaches creates an opportunity to embed modeling as a standard, rather than exceptional, practice in first-in-human drug development.</p><p>The author declares no conflicts of interest.</p><p>No funding was obtained for this work.</p>\",\"PeriodicalId\":10495,\"journal\":{\"name\":\"Clinical Pharmacology in Drug Development\",\"volume\":\"14 10\",\"pages\":\"738-741\"},\"PeriodicalIF\":1.8000,\"publicationDate\":\"2025-10-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://accp1.onlinelibrary.wiley.com/doi/epdf/10.1002/cpdd.1607\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Clinical Pharmacology in Drug Development\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://accp1.onlinelibrary.wiley.com/doi/10.1002/cpdd.1607\",\"RegionNum\":4,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q3\",\"JCRName\":\"PHARMACOLOGY & PHARMACY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Clinical Pharmacology in Drug Development","FirstCategoryId":"3","ListUrlMain":"https://accp1.onlinelibrary.wiley.com/doi/10.1002/cpdd.1607","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"PHARMACOLOGY & PHARMACY","Score":null,"Total":0}
引用次数: 0

摘要

基于模型的药物开发(MIDD)已经从一项有前途的创新发展成为药物开发中的监管要求。过去一年,监管格局发生了变化。美国食品和药物管理局(FDA)在optimus项目下完成了肿瘤剂量优化指南1,2,并将其MIDD配对会议计划制度化3,而ICH发布了用于儿科外推的E11A 4,并发布了由ICH主持制定的关于广义MIDD原则的指南草案M15 5这些进展共同改变了早期药物开发:目标不再仅仅是确定耐受剂量,而是定量地证明关键试验的最佳剂量。通过整合药代动力学(PK)、药效学(PD)、系统药理学和真实世界数据,MIDD降低了开发风险,加速了首次人体研究,并实现了合理的剂量选择。它对决策的影响日益扩大,强调了它不仅是一种技术方法,而且是早期发展战略中的一种范式转变。在过去十年中,由于计算建模的进步和日益有利的监管环境,MIDD的应用得到了显著扩展。FDA和欧洲药品管理局(EMA)已经明确了鼓励在早期开发中使用MIDD的框架,FDA的MIDD试点项目作为促进发起人与监管机构对话的基石倡议。3,5 -7到2025年,这些努力已经成熟,MIDD现在被认为是肿瘤、罕见病和免疫学监管申报的核心。最近的文献强调,MIDD能够以前所未有的精度将临床前数据转化为临床相关的预测。例如,Ren等人强调了PK/PD建模如何在发现和开发中改进剂量优化策略,从而可以更早地确定最佳治疗窗口这在首次人体试验中具有相关性,其中剂量递增策略现在可以通过定量预测而不是经验性的试错来提供信息。这一转变将早期发展从“第一次安全剂量”的思维转变为“第一次科学信息剂量”。首次人体剂量预测。基于生理的药代动力学(PBPK)模型已经彻底改变了首次在人体内的剂量选择,特别是对于具有复杂代谢的生物制剂和小分子。PBPK模型结合了虚拟人群中吸收、代谢和清除的可变性,提供了超越传统异速缩放的机制见解在肿瘤学和罕见疾病中,这种模型越来越多地用于支持起始剂量合理化,最大限度地减少剂量不足和毒性风险。适应性试验设计。MIDD促进了I期研究的适应性试验设计。通过将贝叶斯分层模型与实时PK/PD数据相结合,研究人员可以动态调整剂量递增方案。这减少了试验持续时间和参与者暴露于亚治疗剂量或不安全剂量。FDA最近对适应性MIDD框架的认可强调了其监管接受度。1,3,53。药物-药物相互作用(DDI)风险缓解。PBPK模型现在通常用于预测细胞色素450 (CYP)介导和转运蛋白介导的DDI,从而减少了广泛的临床DDI研究的需要这种集成节省了时间和资源,同时为监管机构提供了机制上的理由。罕见病和精准医学。对于患者数量本来就很少的罕见疾病,MIDD支持从稀疏数据集进行外推,为早期给药策略提供信息。Chen等人(2025)指出,MIDD还为评估癌症中翻译后修饰的药理学调节提供了途径,其中生物标志物驱动的建模对于剂量选择至关重要。人工智能(AI)和机器学习的发展进一步增强了MIDD。这些技术允许使用电子健康记录数据和可穿戴生物标志物实时更新人口PK模型,从而在早期试验期间持续改进模型结合机械PBPK和数据驱动的人工智能模型的混合方法正在成为标准,提高了不同人群的预测准确性。另一个前沿领域是定量系统药理学(QSP)。QSP集成了网络生物学、组学数据和机制建模来理解药物-疾病相互作用。它在I期研究中的应用正在扩大,特别是在免疫肿瘤学和代谢紊乱方面,动态反馈回路使剂量-反应预测复杂化。赞美MIDD概念上的优雅是一回事;显示出切实的回报是另一回事。分析表明,在时间和成本方面都有相当大的节省。 重要的是,这些节省不仅仅是金钱上的或时间上的——它们重塑了战略决策。MIDD促进了早期“不去”的决定,资源的重新分配,并提高了对剂量策略的信心。自动化工具,例如第一阶段的自动化监控;用于I期数据处理的2 (AMP)和用于浓度- qt分析的心脏暴露-反应建模(CardioERM)进一步压缩了时间线,将数周的报告生成缩短为几分钟,每年节省数百个工作日。2025年的监管环境将积极拥抱MIDD,发布关于PBPK、population PK和QSP在早期开发中的使用的详细指导。3,5,6,15,16监管科学的协作性越来越强,有专门讨论模型可信度的研讨会和ind前咨询。重要的是,监管机构现在希望赞助商证明模型如何影响剂量选择和试验设计。从伦理上讲,MIDD减少了不必要的人类接触不安全剂量,符合最小化参与者风险的原则。在罕见病和儿科疾病中,MIDD提供了从成人或临床前数据推断给药方案的伦理途径,减轻了弱势群体的负担。尽管取得了进展,但挑战依然存在。模型的可信度和可重复性仍然令人担忧,特别是在整合人工智能驱动的预测时。建模假设的透明度和严格的资格框架对于维持监管机构和临床医生之间的信任至关重要此外,从临床前模型到现实世界数据集的数据源的异质性带来了需要标准化的集成挑战。MIDD在早期开发中的演变与人工智能和机器学习支持的建模方法的日益采用是平行的。随着真实世界的数据和计算机方法越来越多地整合到临床前到临床的转化中,现在被称为MID3(模型信息药物发现和开发)21的转变变得越来越明显。在这个汇聚点上,对建模专业知识、基础设施和监管行业协作的投资对于维持其影响至关重要。MIDD不仅代表了一个技术工具包,而且代表了早期创新的范式转变。通过整合定量预测、机制建模和适应性学习,MIDD降低了早期临床开发的风险,并为后续阶段建立了更强大的科学基础。监管机构愿意接受这些方法,这为在首次人体药物开发中将建模作为标准而不是例外的实践创造了机会。作者声明无利益冲突。这项工作没有获得资金。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

Model-Informed Drug Development in Early-Phase Development: Navigating Complexity With Quantitative Clarity

Model-Informed Drug Development in Early-Phase Development: Navigating Complexity With Quantitative Clarity

Model-informed drug development (MIDD) has evolved from a promising innovation to a regulatory imperative in drug development. Over the past year, the regulatory landscape shifted. The Food and Drug Administration (FDA) finalized guidance on oncology dose optimization under Project Optimus1, 2 and institutionalized its MIDD Paired Meeting Program,3 while ICH issued E11A for pediatric extrapolation4 and released a draft guideline M15, developed under ICH auspices, on generalized MIDD principles.5 These developments collectively transform early drug development: the goal is no longer just identifying a tolerated dose but quantitatively justifying optimal dosing for pivotal trials. By integrating pharmacokinetics (PK), pharmacodynamics (PD), systems pharmacology, and real-world data, MIDD reduces development risks, accelerates first-in-human studies, and enables rational dose selection. Its expanding influence on decision-making underscores its role not only as a technical methodology but as a paradigm shift in early-phase development strategy.

In the last decade, the utility of MIDD has expanded significantly, driven by advances in computational modeling and an increasingly favorable regulatory environment. The FDA and the European Medicines Agency (EMA) have articulated frameworks encouraging the use of MIDD in early development, with the FDA's MIDD Pilot Program serving as a cornerstone initiative fostering sponsor–regulator dialogue.3, 5-7 In 2025, such efforts have matured, with MIDD now recognized as central to regulatory submissions in oncology, rare diseases, and immunology.

Recent literature emphasizes that MIDD enables the translation of preclinical data into clinically relevant predictions with unprecedented precision. For example, Ren et al8 highlight how PK/PD modeling refines dose optimization strategies in discovery and development, allowing earlier identification of optimal therapeutic windows.3 This has relevance in first-in-human trials, where dose escalation strategies can now be informed by quantitative predictions rather than empirical trial-and-error. This shift reframes early-phase development from “first safe dose” thinking to “first scientifically informative dose.”

1. First-in-Human Dose Prediction. Physiologically based pharmacokinetic (PBPK) modeling has revolutionized first-in-human dose selection, particularly for biologics and small molecules with complex metabolism. PBPK models incorporate variability in absorption, metabolism, and clearance across virtual populations, offering mechanistic insights that go beyond traditional allometric scaling.9 In oncology and rare diseases, such models are increasingly used to support starting dose rationalization, minimizing both underdosing and toxicity risk.

2. Adaptive Trial Designs. MIDD facilitates adaptive trial designs in Phase I studies. By integrating Bayesian hierarchical models with real-time PK/PD data, investigators can adjust dose escalation schemes dynamically. This reduces trial duration and exposure of participants to subtherapeutic or unsafe doses. The FDA's recent endorsements of adaptive MIDD frameworks underscore their regulatory acceptance.1, 3, 5

3. Drug–Drug Interaction (DDI) Risk Mitigation. PBPK models are now routinely leveraged to predict cytochrome-450 (CYP)-mediated and transporter-mediated DDIs early in development, reducing the need for extensive dedicated clinical DDI studies.10 This integration saves time and resources while providing mechanistic justifications to regulators.

4. Rare Disease and Precision Medicine. For rare diseases, where patient numbers are inherently small, MIDD supports extrapolation from sparse datasets to inform early-phase dosing strategies. Chen et al (2025) note that MIDD also provides a path for evaluating pharmacological modulation of post-translational modifications in cancer, where biomarker-driven modeling is critical for dose selection.11

The growth of artificial intelligence (AI) and machine learning has further augmented MIDD. These technologies allow for real-time updating of population PK models with electronic health record data and wearable-derived biomarkers, enabling continuous model refinement during early-phase trials.12 Hybrid approaches that combine mechanistic PBPK with data-driven AI models are becoming the standard, improving predictive accuracy across diverse populations.

Another frontier is quantitative systems pharmacology (QSP). QSP integrates network biology, omics data, and mechanistic modeling to understand drug–disease interactions. Its application in Phase I studies is expanding, particularly in immuno-oncology and metabolic disorders, where dynamic feedback loops complicate dose–response predictions.13

It is one thing to laud MIDD's conceptual elegance; it is another to show tangible returns. Analyses have shown considerable savings both in terms of time and costs. Importantly, these savings are not just monetary or temporal—they reshape strategic decision-making. MIDD facilitates early “No-Go” decisions, reallocation of resources, and heightened confidence in dosing strategy. Automation tools such as Automated Monitoring of Phase I; 2 (AMP) for Phase I data processing and Cardio Exposure–Response Modeling (CardioERM) for concentration–QT analysis further compressed timelines, turning weeks of report generation into minutes and saving hundreds of business days annually.14

The regulatory environment in 2025 actively embraces MIDD, publishing detailed guidance on the use of PBPK, population PK, and QSP in early development.3, 5, 6, 15, 16 Regulatory science is increasingly collaborative, with workshops and pre-IND consultations dedicated to discussing model credibility. Importantly, regulators now expect sponsors to demonstrate how modeling influenced dose selection and trial design.

Ethically, MIDD reduces unnecessary human exposure to unsafe doses, aligning with the principle of minimizing participant risk. In rare and pediatric diseases, MIDD provides an ethical pathway for extrapolating dosing regimens from adult or preclinical data, reducing the burden on vulnerable populations.17

Despite progress, challenges persist. Model credibility and reproducibility remain concerns, particularly when integrating AI-driven predictions. Transparency in modeling assumptions and rigorous qualification frameworks are essential to maintain trust among regulators and clinicians.12 Moreover, the heterogeneity of data sources from preclinical models to real-world datasets poses integration challenges that require standardization.

The evolution of MIDD in early development parallels the growing adoption of AI and machine learning-enabled modeling approaches. As real-world data and in silico methods become increasingly integrated into preclinical-to-clinical translation, a shift toward what is now termed MID3 (model-informed drug discovery and development)21 is becoming evident. At this convergence point, investment in modeling expertise, infrastructure, and regulatory–industry collaboration is essential to sustain its impact.

MIDD represents not merely a technical toolkit but a paradigm shift for early-phase innovation. By integrating quantitative predictions, mechanistic modeling, and adaptive learning, MIDD de-risks early clinical development and establishes a stronger scientific foundation for subsequent phases. Regulatory willingness to embrace these approaches creates an opportunity to embed modeling as a standard, rather than exceptional, practice in first-in-human drug development.

The author declares no conflicts of interest.

No funding was obtained for this work.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
3.70
自引率
10.00%
发文量
154
期刊介绍: Clinical Pharmacology in Drug Development is an international, peer-reviewed, online publication focused on publishing high-quality clinical pharmacology studies in drug development which are primarily (but not exclusively) performed in early development phases in healthy subjects.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信