Model-Informed Drug Development: Bang for the Buck?

IF 5.5 2区 医学 Q1 PHARMACOLOGY & PHARMACY
Allison Dunn, Piet H. van der Graaf
{"title":"Model-Informed Drug Development: Bang for the Buck?","authors":"Allison Dunn,&nbsp;Piet H. van der Graaf","doi":"10.1002/cpt.3744","DOIUrl":null,"url":null,"abstract":"<p>Model-Informed Drug Development (MIDD) has emerged as a foundational pillar of modern drug development, transforming how evidence is generated, integrated, and acted upon throughout the lifecycle. Once viewed as a complementary strategy, MIDD is now routinely embedded in regulatory and industry decision-making, offering powerful tools to optimize study design, inform dose selection, and support benefit–risk assessments. Its impact is both measurable, saving time and reducing costs by streamlining development, and qualitative, reflecting the critical, but less easily quantified, value of more informed labeling statements that support clinical decision-making and patient care. As MIDD continues to evolve, its influence can be understood across three core areas: (1) improving efficiency and generating cost savings, (2) mitigating risk across development programs, and (3) enhancing product labeling to inform real-world decisions. Together, these dimensions illustrate the value of MIDD not only as a technical approach, but also as a strategic framework central to the future of drug development.</p><p>One of the most evident and widely recognized impacts of MIDD is its ability to improve the efficiency and reduce the overall cost of drug development. By integrating quantitative models early and throughout the development process, MIDD enables more strategic decision-making, allowing sponsors to streamline programs, reduce redundancy, and target resources more effectively. For example, model-based bridging approaches can support smaller or fewer clinical trials by extrapolating existing data across populations or dosing regimens, while optimized dose selection reduces the risk of trial failure due to suboptimal exposure-response relationships. Moreover, model-based trial designs, including adaptive designs, can accelerate Investigational New Drug and New Drug Application timelines by informing dose-ranging studies, refining endpoints, or enabling innovative approaches such as dose-exposure extrapolation or the waiver of confirmatory trials in specific contexts.</p><p>These methodological efficiencies are translating into tangible economic value. In this issue of <i>Clinical Pharmacology &amp; Therapeutics</i> (<i>CPT</i>), a recent analysis by Pfizer found that the use of MIDD approaches was associated with an average reduction of 10 months in development cycle time and $5 million in development costs per program.<span><sup>1</sup></span> To the best of our knowledge, this is the first retrospective analysis of internal research and development (R&amp;D) programs quantifying the broader organizational value of MIDD by capturing the key development questions informed by MIDD, associated assumptions and risks, and the potential impact on cost, timelines, and decision-making. Methods based on per-subject approximations and trial size reductions were used to estimate cost and time savings, using benchmarks for study timelines and enrollment metrics across development phases. This work expands upon a 2013 <i>CPT</i> publication from Pfizer which first highlighted the potential for model-based strategies to generate cost savings in the range of $70 million per year.<span><sup>2</sup></span> Another prior analysis found that MIDD implementation can reduce development costs by $30 to $70 million, depending on the scope and integration of modeling approaches.<span><sup>3</sup></span> These benefits reflect not only streamlined development timelines but also earlier and more confident go/no-go decisions that can be enabled by predictive simulations. This ever-evolving role of MIDD as a driver of efficient development is also highlighted on the cover of this issue (<b>Figure</b> 1).</p><p>Importantly, the economic benefits of MIDD extend beyond the point of regulatory approval. Traditionally, methods such as model-based meta-analysis (MBMA) have been known to strengthen the quality of clinical evidence. In one article in this issue of <i>CPT</i>, a less common application is highlighted, demonstrating how this methodology was used to improve the likelihood of favorable reimbursement decisions and facilitates more timely access for patients to innovative therapies.<span><sup>4</sup></span> In this way, MIDD supports not only efficient development, but also more efficient delivery of therapeutic value to the healthcare system.</p><p>MIDD also plays a critical role in mitigating risk throughout the drug development lifecycle by leveraging computational tools and predictive models to optimize decision-making. This approach helps reduce the likelihood of failure and unforeseen complications, particularly as drug programs move from early to late-stage development. One of the primary applications of MIDD is in optimizing dose selection, directly addressing the risk of advancing a development program with suboptimal efficacy and/or safety profiles. Using population pharmacokinetic (popPK) modeling and exposure-response (ER) assessments, MIDD enables the identification of an optimal dose range that maximizes therapeutic benefits while minimizing adverse effects. This improves the likelihood of success in clinical trials, reducing both time and costs associated with development. Beyond dose optimization, MIDD also aids in forecasting the probability of technical, commercial, and regulatory success throughout the development process. By quantitatively assessing the likelihood of success at various stages, MIDD helps prioritize resources and focus efforts on high-probability programs, thus minimizing investments in projects with lower chances of success. In an environment where the costs of failure are high, this potential to assess and mitigate risk early in development is invaluable.</p><p>MIDD is especially valuable in mitigating risk for novel therapeutic modalities, where traditional approaches may be limited due to scientific uncertainty or lack of historical data. In these cases, predictive modeling can also guide the selection of dosing regimens and anticipate potential clinical outcomes. For example, in this issue of <i>CPT</i>, the FDA describes its experience with the development of chimeric antigen receptor (CAR) T-cell therapies, a class of “living drugs.” They focus on how the unique pharmacokinetic and pharmacodynamic behaviors present significant challenges for conventional methods.<span><sup>5</sup></span> Learnings from the FDA demonstrate how popPK and ER modeling can be readily leveraged to overcome these challenges, particularly for dose selection and ensuring internal consistency of clinical data. Similarly, this issue of <i>CPT</i> explores how novel modalities like bispecific antibodies present their own set of challenges due to their unique mechanism of action.<span><sup>6</sup></span> These antibodies bind to two distinct targets <i>in vivo</i>, forming a trimolecular complex that often results in a bell-shaped concentration-response curve. Traditional approaches may struggle to address these complexities, but the application of mechanistic physiologically based pharmacokinetic (PBPK) and response-optimized models enables more accurate predictions of clinical outcomes. These examples showcase how MIDD plays a pivotal role in mitigating risks throughout the drug development process, particularly for innovative and complex therapeutic modalities, by enabling more informed decision-making and optimization strategies.</p><p>MIDD has had a transformative impact on the way drug labeling supports clinical decision-making, particularly when it comes to dose optimization for specific patient populations. In the past, labeling often included generalized dosing recommendations that may not have initially accounted for the variability seen across different demographic groups, such as older adults, pregnant individuals, or pediatric patients. However, with the rise of MIDD, the ability to generate more informative and personalized labeling statements has been greatly enhanced, ensuring that the right drug, at the right dose, reaches the right patient.</p><p>One of the core values of MIDD lies in its patient-centricity. By utilizing advanced modeling approaches, MIDD facilitates more accurate dose selection, particularly for populations where clinical trial data may be challenging to generate. For example, the application of PBPK modeling has been instrumental in supporting dose recommendations for pregnant individuals, a group traditionally excluded from clinical trials. One article in this issue of <i>CPT</i> explores its application to therapeutic antibodies, which are often prescribed off-label during pregnancy, and shows how PBPK modeling can be used to assess drug exposure and guide dose adjustments in this population.<span><sup>7</sup></span> Similarly, MIDD has proven invaluable in pediatric drug development, where dosing regimens often rely on extrapolations from adult data. Several studies in this area have utilized modeling and simulation to refine pediatric dosing recommendations. For instance, two articles in the current issue describe how MIDD supported apixaban dosing in pediatric patients, helping to optimize the therapeutic regimen while minimizing adverse effects.<span><sup>8, 9</sup></span> In another example, popPK and ER modeling was used to optimize the dose of Brentuximab Vedotin in pediatric patients with advanced-stage newly diagnosed Hodgkin lymphoma.<span><sup>10</sup></span> These cases demonstrate how MIDD contributes to better clinical decision-making by improving the precision and accuracy of labeling information. The ability to tailor dosing recommendations to specific populations is a critical advancement that supports more effective and safer treatments, ultimately benefiting both patients and healthcare providers.</p><p>Today, MIDD is considered a mainstay of modern development efforts. As the landscape of drug development continues to evolve, the future of MIDD lies in the integration of more advanced methodologies, particularly artificial intelligence (AI), machine learning (ML) and real-world data, across the entire development continuum. This vision aligns with a broader reframing of MIDD as MID3: Model-Informed Drug <i>Discovery</i> and Development. For example, AI and ML approaches can be used early in drug discovery to identify novel targets, predict compound activity, and prioritize candidates with favorable safety and efficacy profiles. Modeling and simulation methodologies play a critical role in every facet of development and regulatory decision-making, and the integration of AI/ML across this landscape has the potential to further strengthen these efforts.</p><p>Recent FDA draft guidance recognizes the potential of AI/ML in regulatory decision-making, signaling an increasing openness from regulatory bodies to embrace these technologies.<span><sup>11</sup></span> Early engagement from these agencies underscores the importance of staying ahead of technological advancements and leveraging them to enhance MIDD practices. While recent analyses, such as Pfizer's report in this issue of CPT that estimates cost savings associated with MIDD strategies,<span><sup>1</sup></span> have helped quantify the downstream impact of model-informed approaches, future work may further expand on this foundation by exploring the potential value of modeling and AI/ML in preclinical and discovery phases. For instance, AI-enabled target identification, lead optimization, and early safety prediction could generate additional efficiencies and cost savings upstream, further strengthening the overall case for investment in MID3.</p><p>Looking ahead, AI/ML techniques are likely to be incorporated into every stage of the development lifecycle. In the case of drug discovery, AI/ML can accelerate identification of effective therapies by enabling more precise target identification and validation. By leveraging data sources, such as multi-omic repositories and extensive literature-based datasets, these technologies can uncover novel disease targets and prioritize those with the highest translational potential. In clinical development, AI-driven patient stratification and trial enrichment will identify subpopulations most likely to respond to therapies, facilitating shorter trials with higher success rates. On the operational side, AI has the potential to automate clinical trial monitoring, streamlining data review, detecting safety signals in real-time, and dynamically adjusting trial protocols as needed, which could substantially reduce both operational costs and risks. Moreover, AI-enabled systems, such as digital twins, can simulate virtual populations and predict trial outcomes, providing invaluable support for regulatory submissions and post-market commitments. Post-market surveillance could likewise benefit from AI-powered pharmacovigilance systems, which can analyze vast amounts of real-world data, ranging from EHRs to social media and spontaneous reporting systems, to detect adverse event patterns more quickly and accurately than traditional methods.</p><p>In light of these advancements, the call for broader adoption and continued training in AI/ML tools within drug development is paramount. As the integration of these technologies becomes increasingly central to the drug development process, it is crucial for stakeholders to invest in the necessary infrastructure, training, and interdisciplinary collaborations to ensure that these tools are appropriately incorporated into a development program. Just as a concerted investment in pharmacometrics laid the groundwork for the rise of MIDD, we now stand at a similar inflection point where embracing AI/ML with the same level of commitment can redefine what is possible in the next era of drug development.</p><p>No funding was received for this work.</p><p>The authors declared no competing interests for this work.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":"118 2","pages":"283-287"},"PeriodicalIF":5.5000,"publicationDate":"2025-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cpt.3744","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Clinical Pharmacology & Therapeutics","FirstCategoryId":"3","ListUrlMain":"https://ascpt.onlinelibrary.wiley.com/doi/10.1002/cpt.3744","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"PHARMACOLOGY & PHARMACY","Score":null,"Total":0}
引用次数: 0

Abstract

Model-Informed Drug Development (MIDD) has emerged as a foundational pillar of modern drug development, transforming how evidence is generated, integrated, and acted upon throughout the lifecycle. Once viewed as a complementary strategy, MIDD is now routinely embedded in regulatory and industry decision-making, offering powerful tools to optimize study design, inform dose selection, and support benefit–risk assessments. Its impact is both measurable, saving time and reducing costs by streamlining development, and qualitative, reflecting the critical, but less easily quantified, value of more informed labeling statements that support clinical decision-making and patient care. As MIDD continues to evolve, its influence can be understood across three core areas: (1) improving efficiency and generating cost savings, (2) mitigating risk across development programs, and (3) enhancing product labeling to inform real-world decisions. Together, these dimensions illustrate the value of MIDD not only as a technical approach, but also as a strategic framework central to the future of drug development.

One of the most evident and widely recognized impacts of MIDD is its ability to improve the efficiency and reduce the overall cost of drug development. By integrating quantitative models early and throughout the development process, MIDD enables more strategic decision-making, allowing sponsors to streamline programs, reduce redundancy, and target resources more effectively. For example, model-based bridging approaches can support smaller or fewer clinical trials by extrapolating existing data across populations or dosing regimens, while optimized dose selection reduces the risk of trial failure due to suboptimal exposure-response relationships. Moreover, model-based trial designs, including adaptive designs, can accelerate Investigational New Drug and New Drug Application timelines by informing dose-ranging studies, refining endpoints, or enabling innovative approaches such as dose-exposure extrapolation or the waiver of confirmatory trials in specific contexts.

These methodological efficiencies are translating into tangible economic value. In this issue of Clinical Pharmacology & Therapeutics (CPT), a recent analysis by Pfizer found that the use of MIDD approaches was associated with an average reduction of 10 months in development cycle time and $5 million in development costs per program.1 To the best of our knowledge, this is the first retrospective analysis of internal research and development (R&D) programs quantifying the broader organizational value of MIDD by capturing the key development questions informed by MIDD, associated assumptions and risks, and the potential impact on cost, timelines, and decision-making. Methods based on per-subject approximations and trial size reductions were used to estimate cost and time savings, using benchmarks for study timelines and enrollment metrics across development phases. This work expands upon a 2013 CPT publication from Pfizer which first highlighted the potential for model-based strategies to generate cost savings in the range of $70 million per year.2 Another prior analysis found that MIDD implementation can reduce development costs by $30 to $70 million, depending on the scope and integration of modeling approaches.3 These benefits reflect not only streamlined development timelines but also earlier and more confident go/no-go decisions that can be enabled by predictive simulations. This ever-evolving role of MIDD as a driver of efficient development is also highlighted on the cover of this issue (Figure 1).

Importantly, the economic benefits of MIDD extend beyond the point of regulatory approval. Traditionally, methods such as model-based meta-analysis (MBMA) have been known to strengthen the quality of clinical evidence. In one article in this issue of CPT, a less common application is highlighted, demonstrating how this methodology was used to improve the likelihood of favorable reimbursement decisions and facilitates more timely access for patients to innovative therapies.4 In this way, MIDD supports not only efficient development, but also more efficient delivery of therapeutic value to the healthcare system.

MIDD also plays a critical role in mitigating risk throughout the drug development lifecycle by leveraging computational tools and predictive models to optimize decision-making. This approach helps reduce the likelihood of failure and unforeseen complications, particularly as drug programs move from early to late-stage development. One of the primary applications of MIDD is in optimizing dose selection, directly addressing the risk of advancing a development program with suboptimal efficacy and/or safety profiles. Using population pharmacokinetic (popPK) modeling and exposure-response (ER) assessments, MIDD enables the identification of an optimal dose range that maximizes therapeutic benefits while minimizing adverse effects. This improves the likelihood of success in clinical trials, reducing both time and costs associated with development. Beyond dose optimization, MIDD also aids in forecasting the probability of technical, commercial, and regulatory success throughout the development process. By quantitatively assessing the likelihood of success at various stages, MIDD helps prioritize resources and focus efforts on high-probability programs, thus minimizing investments in projects with lower chances of success. In an environment where the costs of failure are high, this potential to assess and mitigate risk early in development is invaluable.

MIDD is especially valuable in mitigating risk for novel therapeutic modalities, where traditional approaches may be limited due to scientific uncertainty or lack of historical data. In these cases, predictive modeling can also guide the selection of dosing regimens and anticipate potential clinical outcomes. For example, in this issue of CPT, the FDA describes its experience with the development of chimeric antigen receptor (CAR) T-cell therapies, a class of “living drugs.” They focus on how the unique pharmacokinetic and pharmacodynamic behaviors present significant challenges for conventional methods.5 Learnings from the FDA demonstrate how popPK and ER modeling can be readily leveraged to overcome these challenges, particularly for dose selection and ensuring internal consistency of clinical data. Similarly, this issue of CPT explores how novel modalities like bispecific antibodies present their own set of challenges due to their unique mechanism of action.6 These antibodies bind to two distinct targets in vivo, forming a trimolecular complex that often results in a bell-shaped concentration-response curve. Traditional approaches may struggle to address these complexities, but the application of mechanistic physiologically based pharmacokinetic (PBPK) and response-optimized models enables more accurate predictions of clinical outcomes. These examples showcase how MIDD plays a pivotal role in mitigating risks throughout the drug development process, particularly for innovative and complex therapeutic modalities, by enabling more informed decision-making and optimization strategies.

MIDD has had a transformative impact on the way drug labeling supports clinical decision-making, particularly when it comes to dose optimization for specific patient populations. In the past, labeling often included generalized dosing recommendations that may not have initially accounted for the variability seen across different demographic groups, such as older adults, pregnant individuals, or pediatric patients. However, with the rise of MIDD, the ability to generate more informative and personalized labeling statements has been greatly enhanced, ensuring that the right drug, at the right dose, reaches the right patient.

One of the core values of MIDD lies in its patient-centricity. By utilizing advanced modeling approaches, MIDD facilitates more accurate dose selection, particularly for populations where clinical trial data may be challenging to generate. For example, the application of PBPK modeling has been instrumental in supporting dose recommendations for pregnant individuals, a group traditionally excluded from clinical trials. One article in this issue of CPT explores its application to therapeutic antibodies, which are often prescribed off-label during pregnancy, and shows how PBPK modeling can be used to assess drug exposure and guide dose adjustments in this population.7 Similarly, MIDD has proven invaluable in pediatric drug development, where dosing regimens often rely on extrapolations from adult data. Several studies in this area have utilized modeling and simulation to refine pediatric dosing recommendations. For instance, two articles in the current issue describe how MIDD supported apixaban dosing in pediatric patients, helping to optimize the therapeutic regimen while minimizing adverse effects.8, 9 In another example, popPK and ER modeling was used to optimize the dose of Brentuximab Vedotin in pediatric patients with advanced-stage newly diagnosed Hodgkin lymphoma.10 These cases demonstrate how MIDD contributes to better clinical decision-making by improving the precision and accuracy of labeling information. The ability to tailor dosing recommendations to specific populations is a critical advancement that supports more effective and safer treatments, ultimately benefiting both patients and healthcare providers.

Today, MIDD is considered a mainstay of modern development efforts. As the landscape of drug development continues to evolve, the future of MIDD lies in the integration of more advanced methodologies, particularly artificial intelligence (AI), machine learning (ML) and real-world data, across the entire development continuum. This vision aligns with a broader reframing of MIDD as MID3: Model-Informed Drug Discovery and Development. For example, AI and ML approaches can be used early in drug discovery to identify novel targets, predict compound activity, and prioritize candidates with favorable safety and efficacy profiles. Modeling and simulation methodologies play a critical role in every facet of development and regulatory decision-making, and the integration of AI/ML across this landscape has the potential to further strengthen these efforts.

Recent FDA draft guidance recognizes the potential of AI/ML in regulatory decision-making, signaling an increasing openness from regulatory bodies to embrace these technologies.11 Early engagement from these agencies underscores the importance of staying ahead of technological advancements and leveraging them to enhance MIDD practices. While recent analyses, such as Pfizer's report in this issue of CPT that estimates cost savings associated with MIDD strategies,1 have helped quantify the downstream impact of model-informed approaches, future work may further expand on this foundation by exploring the potential value of modeling and AI/ML in preclinical and discovery phases. For instance, AI-enabled target identification, lead optimization, and early safety prediction could generate additional efficiencies and cost savings upstream, further strengthening the overall case for investment in MID3.

Looking ahead, AI/ML techniques are likely to be incorporated into every stage of the development lifecycle. In the case of drug discovery, AI/ML can accelerate identification of effective therapies by enabling more precise target identification and validation. By leveraging data sources, such as multi-omic repositories and extensive literature-based datasets, these technologies can uncover novel disease targets and prioritize those with the highest translational potential. In clinical development, AI-driven patient stratification and trial enrichment will identify subpopulations most likely to respond to therapies, facilitating shorter trials with higher success rates. On the operational side, AI has the potential to automate clinical trial monitoring, streamlining data review, detecting safety signals in real-time, and dynamically adjusting trial protocols as needed, which could substantially reduce both operational costs and risks. Moreover, AI-enabled systems, such as digital twins, can simulate virtual populations and predict trial outcomes, providing invaluable support for regulatory submissions and post-market commitments. Post-market surveillance could likewise benefit from AI-powered pharmacovigilance systems, which can analyze vast amounts of real-world data, ranging from EHRs to social media and spontaneous reporting systems, to detect adverse event patterns more quickly and accurately than traditional methods.

In light of these advancements, the call for broader adoption and continued training in AI/ML tools within drug development is paramount. As the integration of these technologies becomes increasingly central to the drug development process, it is crucial for stakeholders to invest in the necessary infrastructure, training, and interdisciplinary collaborations to ensure that these tools are appropriately incorporated into a development program. Just as a concerted investment in pharmacometrics laid the groundwork for the rise of MIDD, we now stand at a similar inflection point where embracing AI/ML with the same level of commitment can redefine what is possible in the next era of drug development.

No funding was received for this work.

The authors declared no competing interests for this work.

Abstract Image

Abstract Image

Abstract Image

Abstract Image

基于模型的药物开发:物有所值?
模型知情药物开发(MIDD)已成为现代药物开发的基础支柱,改变了整个生命周期中证据的生成、整合和作用方式。MIDD曾被视为一种补充策略,现在已常规地纳入监管和行业决策,为优化研究设计、告知剂量选择和支持获益风险评估提供了强大的工具。它的影响是可衡量的,通过简化开发节省时间和降低成本,并且是定性的,反映了支持临床决策和患者护理的更知情的标签声明的关键但不易量化的价值。随着MIDD的不断发展,它的影响可以在三个核心领域得到理解:(1)提高效率和节省成本,(2)降低开发项目的风险,(3)增强产品标签,为现实世界的决策提供信息。总之,这些方面说明了MIDD不仅作为一种技术方法,而且作为对药物开发未来至关重要的战略框架的价值。MIDD最明显和广泛认可的影响之一是它能够提高效率并降低药物开发的总体成本。通过在早期和整个开发过程中集成定量模型,MIDD支持更多的战略决策,允许发起人简化程序,减少冗余,并更有效地定位资源。例如,基于模型的桥接方法可以通过在人群或给药方案中推断现有数据来支持更小或更少的临床试验,而优化的剂量选择可以降低由于次优暴露-反应关系而导致试验失败的风险。此外,基于模型的试验设计,包括自适应设计,可以通过通知剂量范围研究,改进终点或启用创新方法(如剂量暴露外推法或在特定情况下放弃验证性试验)来加快新药研究和新药申请的时间表。这些方法上的效率正在转化为有形的经济价值。在这一期的《临床药理学》杂志上;辉瑞公司最近的一项分析发现,使用MIDD方法与平均减少10个月的开发周期时间和每个项目500万美元的开发成本有关据我们所知,这是对内部研究和开发(R&amp;D)计划的第一次回顾性分析,通过捕获由MIDD通知的关键开发问题、相关的假设和风险,以及对成本、时间线和决策的潜在影响,量化了MIDD的更广泛的组织价值。使用基于每个受试者近似值和试验规模缩减的方法来估计成本和时间节省,使用跨开发阶段的研究时间表和入组指标的基准。这项工作扩展了辉瑞公司2013年的CPT出版物,该出版物首次强调了基于模型的策略每年可节省7000万美元的成本另一个先前的分析发现,MIDD实现可以减少3000万到7000万美元的开发成本,这取决于建模方法的范围和集成这些好处不仅反映了简化的开发时间表,而且可以通过预测模拟实现更早和更自信的决定。作为高效开发的驱动因素,MIDD不断发展的角色也在本期封面上得到了强调(图1)。重要的是,MIDD的经济效益超出了监管机构批准的范围。传统上,诸如基于模型的荟萃分析(MBMA)等方法已被认为可以提高临床证据的质量。在本期《CPT》的一篇文章中,强调了一种不太常见的应用,展示了如何使用这种方法来提高有利的报销决策的可能性,并促进患者更及时地获得创新疗法通过这种方式,MIDD不仅支持有效的开发,而且还支持更有效地向医疗保健系统提供治疗价值。MIDD还通过利用计算工具和预测模型来优化决策,在降低整个药物开发生命周期的风险方面发挥着关键作用。这种方法有助于减少失败的可能性和不可预见的并发症,特别是当药物项目从早期发展到后期时。MIDD的主要应用之一是优化剂量选择,直接解决推进具有次优疗效和/或安全性的开发计划的风险。利用群体药代动力学(popPK)模型和暴露-反应(ER)评估,MIDD能够确定最佳剂量范围,最大限度地提高治疗效益,同时最大限度地减少不良反应。 这提高了临床试验成功的可能性,减少了与开发相关的时间和成本。除了剂量优化,MIDD还有助于预测整个开发过程中技术、商业和监管成功的可能性。通过在各个阶段定量地评估成功的可能性,MIDD帮助确定资源的优先级,并将努力集中在高概率的计划上,从而最小化对成功机会较低的项目的投资。在一个失败成本很高的环境中,这种在开发早期评估和减轻风险的潜力是无价的。MIDD在降低新治疗方式的风险方面尤其有价值,传统方法可能由于科学的不确定性或缺乏历史数据而受到限制。在这些情况下,预测模型也可以指导给药方案的选择和预测潜在的临床结果。例如,在本期《CPT》中,FDA描述了其开发嵌合抗原受体(CAR) t细胞疗法的经验,这是一类“活药”。他们关注独特的药代动力学和药效学行为如何对传统方法提出重大挑战FDA的经验表明,popPK和ER模型可以很容易地克服这些挑战,特别是在剂量选择和确保临床数据内部一致性方面。同样,这期《CPT》探讨了双特异性抗体等新模式如何由于其独特的作用机制而呈现出自己的一套挑战这些抗体在体内结合两个不同的靶标,形成一个三分子复合物,通常导致钟形浓度-反应曲线。传统方法可能难以解决这些复杂性,但基于机械生理学的药代动力学(PBPK)和反应优化模型的应用可以更准确地预测临床结果。这些例子展示了MIDD如何在整个药物开发过程中发挥关键作用,特别是对于创新和复杂的治疗模式,通过实现更明智的决策和优化策略。MIDD对药物标签支持临床决策的方式产生了变革性影响,特别是在涉及特定患者群体的剂量优化时。在过去,标签通常包括广义的剂量建议,这些建议最初可能没有考虑到不同人口统计学群体(如老年人、孕妇或儿科患者)的差异。然而,随着MIDD的兴起,生成更多信息和个性化标签声明的能力大大增强,确保正确的药物以正确的剂量到达正确的患者手中。MIDD的核心价值之一就是以患者为中心。通过利用先进的建模方法,MIDD有助于更准确的剂量选择,特别是对于临床试验数据可能难以生成的人群。例如,PBPK模型的应用有助于支持孕妇的剂量建议,这一群体传统上被排除在临床试验之外。本期《CPT》的一篇文章探讨了其在治疗性抗体中的应用,这些抗体通常是在怀孕期间开出的,并展示了PBPK模型如何用于评估该人群的药物暴露和指导剂量调整同样,MIDD已被证明在儿科药物开发中是无价的,在儿科药物开发中,给药方案往往依赖于成人数据的推断。该领域的几项研究利用建模和模拟来完善儿科剂量建议。例如,本期杂志上的两篇文章描述了MIDD如何支持儿科患者使用阿哌沙班,帮助优化治疗方案,同时最大限度地减少不良反应。在另一个例子中,popPK和ER模型被用于优化布伦妥昔单抗韦多汀对晚期新诊断霍奇金淋巴瘤儿童患者的剂量这些病例证明了MIDD如何通过提高标签信息的精确性和准确性来促进更好的临床决策。针对特定人群量身定制剂量建议的能力是一项关键的进步,它支持更有效和更安全的治疗,最终使患者和医疗保健提供者都受益。今天,MIDD被认为是现代开发工作的支柱。随着药物开发领域的不断发展,MIDD的未来取决于在整个开发连续体中整合更先进的方法,特别是人工智能(AI)、机器学习(ML)和现实世界数据。这一愿景与MIDD的更广泛的重构相一致,即MID3:基于模型的药物发现和开发。 例如,人工智能和机器学习方法可用于药物发现的早期,以确定新靶点,预测化合物活性,并优先考虑具有良好安全性和有效性的候选药物。建模和仿真方法在开发和监管决策的各个方面都发挥着关键作用,人工智能/机器学习在这一领域的整合有可能进一步加强这些努力。最近的FDA指南草案认识到AI/ML在监管决策中的潜力,标志着监管机构越来越开放地接受这些技术这些机构的早期参与强调了保持技术进步的领先地位并利用它们来加强MIDD实践的重要性。虽然最近的分析,如辉瑞公司在本期CPT上的报告估计了与MIDD策略相关的成本节约,1已经帮助量化了模型知情方法的下游影响,但未来的工作可能会在此基础上进一步扩展,探索建模和AI/ML在临床前和发现阶段的潜在价值。例如,人工智能支持的目标识别、先导物优化和早期安全预测可以提高上游的效率和成本节约,进一步加强MID3投资的整体理由。展望未来,AI/ML技术可能会被整合到开发生命周期的每个阶段。在药物发现方面,AI/ML可以通过更精确的靶点识别和验证来加速有效疗法的识别。通过利用数据源,如多基因组库和广泛的基于文献的数据集,这些技术可以发现新的疾病靶点,并优先考虑具有最高转化潜力的靶点。在临床开发中,人工智能驱动的患者分层和试验丰富将确定最有可能对治疗产生反应的亚群,促进更短的试验时间和更高的成功率。在操作方面,人工智能有可能自动化临床试验监测,简化数据审查,实时检测安全信号,并根据需要动态调整试验方案,这可以大大降低运营成本和风险。此外,人工智能支持的系统,如数字双胞胎,可以模拟虚拟人群并预测试验结果,为监管提交和上市后承诺提供宝贵的支持。上市后监测同样可以受益于人工智能驱动的药物警戒系统,该系统可以分析从电子病历到社交媒体和自发报告系统的大量真实数据,以比传统方法更快、更准确地检测不良事件模式。鉴于这些进步,呼吁在药物开发中更广泛地采用和持续培训AI/ML工具是至关重要的。随着这些技术的整合在药物开发过程中变得越来越重要,利益相关者必须投资于必要的基础设施、培训和跨学科合作,以确保将这些工具适当地纳入开发计划。正如对药物计量学的一致投资为MIDD的兴起奠定了基础一样,我们现在站在一个类似的拐点上,以同样的承诺拥抱AI/ML可以重新定义下一个药物开发时代的可能性。这项工作没有收到任何资金。作者声明这项工作没有竞争利益。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
CiteScore
12.70
自引率
7.50%
发文量
290
审稿时长
2 months
期刊介绍: Clinical Pharmacology & Therapeutics (CPT) is the authoritative cross-disciplinary journal in experimental and clinical medicine devoted to publishing advances in the nature, action, efficacy, and evaluation of therapeutics. CPT welcomes original Articles in the emerging areas of translational, predictive and personalized medicine; new therapeutic modalities including gene and cell therapies; pharmacogenomics, proteomics and metabolomics; bioinformation and applied systems biology complementing areas of pharmacokinetics and pharmacodynamics, human investigation and clinical trials, pharmacovigilence, pharmacoepidemiology, pharmacometrics, and population pharmacology.
×
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