应对精准给药挑战的最新进展。

IF 6.3 2区 医学 Q1 PHARMACOLOGY & PHARMACY
Iris K. Minichmayr, Tomoyuki Mizuno, Srijib Goswami, Richard W. Peck, Thomas M. Polasek, the American Society of Clinical Pharmacology and Therapeutics Precision Dosing Community
{"title":"应对精准给药挑战的最新进展。","authors":"Iris K. Minichmayr,&nbsp;Tomoyuki Mizuno,&nbsp;Srijib Goswami,&nbsp;Richard W. Peck,&nbsp;Thomas M. Polasek,&nbsp;the American Society of Clinical Pharmacology and Therapeutics Precision Dosing Community","doi":"10.1002/cpt.3365","DOIUrl":null,"url":null,"abstract":"<p>Precision dosing can improve drug therapy in complex, critically ill, and chronically ill patient populations who exhibit vast interindividual variabilities in exposures and responses. This perspective outlines five major challenges in precision dosing and highlights their recent progress: (1) application in drug development, (2) improved clinical trials, (3) usefulness of response biomarkers, (4) confidence in model-informed precision dosing, and (5) receptiveness in clinical practice. Many outstanding opportunities in precision dosing remain within regulatory frameworks, data integration and protection, ethics, and reimbursement strategies.</p><p>Precision dosing is a foundational pillar of personalized medicine. The term has been applied to different dosing strategies aimed at improving outcomes and safety in complex patients displaying substantial interindividual variability in drug exposure and response. Depending on their degree of individualization, various “precision dosing methods” result in dosing recommendations for patient subgroups based on specific indications, genotypes, or patient characteristics like renal function, and/or in more individualized dosing based on measurements obtained during drug treatment, for example, drug concentrations from therapeutic drug monitoring (TDM). In complex situations in which target effect measurement and data interpretation are not straightforward, multiple pieces rather than a single pieces of information are ideally combined for precision dosing, requiring sophisticated predictive models.</p><p>Over the past 5–10 years, precision dosing activities, ranging from dried blood spot analyses of plasma drug concentrations to novel models and educational initiatives fostering better collaboration, have surged, with the goals of benefiting patients, aiding prescribers, saving money, and supporting the development of difficult-to-use drugs. This perspective describes recent advances that address five major challenges of precision dosing (<b>Figure</b> 1), followed by a summary of other challenges with slow progress.</p><p>There are strong economic and logistical reasons to limit the number of test doses studied in clinical development. Despite these challenges, precision dosing may be invaluable in tackling complex drug development scenarios, such as for (1) drugs with narrow therapeutic windows, (2) drugs that can lead to severe adverse effects at otherwise typical doses, (3) drugs used for conditions like cancer that carry serious consequences if undertreated, (4) drugs for treating rare, severe, and progressive diseases, and (5) drugs requiring invasive administration routes, for example, intravitreal or intrathecal.<span><sup>1</sup></span></p><p>Regulatory support is variable, but a successful example of pharma-regulatory collaboration supporting precision dosing is MyPKFit®, a clinical decision support (CDS) tool approved alongside recombinant factor VIII that personalizes dosing in hemophilia A. Also, the ophthalmologic drug faricimab was developed with a precision dosing arm in all four of its pivotal clinical trials, covering two indications, that is, neovascular age-related macular degeneration and diabetic macular edema. Personalized treatment with faricimab was approved, with dosing interval individualization according to disease activity such as visual outcomes.<span><sup>1</sup></span></p><p>Generating clinical trial evidence for precision dosing poses a significant challenge with much progress in recent years. As confidence in dose predictions increases (Challenge 4), incorporating MIPD into prospective clinical trials, such as concentration-controlled or effect-controlled trials, is becoming more common.</p><p>Phase II and III studies with individualized dosing arms rather than fixed dosing arms facilitate the exploration of wider and more nuanced doses, resulting in the approval of dose ranges, and subsequently reduced necessity for off-label dosing. Clinical trials of GLP-1 agonists use this approach because there is a fine balance between efficacy and gastrointestinal intolerance.<span><sup>2</sup></span></p><p>In 2021, the FDA's Oncology Center of Excellence initiated Project Optimus to reform the dose selection paradigm in oncology drug development. Project Optimus's primary goal is to ensure that new cancer drugs are developed with optimal dosing strategies by leveraging modern clinical trial designs and more adaptable, patient-centered approaches to dosing – instead of the conventional maximum tolerated dose approach – that consider individual variability and the unique characteristics of the respective cancer type.</p><p>Substantial portions of pharmacokinetic-pharmacodynamic (PK-PD) variability remain unknown for many drugs. Efforts to further understand disease heterogeneity and variability in drug responses, even among patients with comparable drug exposures, have significantly increased in the last decade with the discovery of novel biomarkers and the development of new, more sensitive analysis methods.</p><p>For certain drugs, TDM utilizing microsampling devices – allowing dried blood spot collection at home and delivery to a laboratory for concentration testing – is feasible.<span><sup>3</sup></span> Furthermore, recent advances in biosensor technology have improved the ability to measure drug concentrations directly and non-invasively.<span><sup>4</sup></span> This extends to biomarker monitoring, exemplified by the boom in continuous blood glucose monitoring patches that provide feedback on antidiabetic drug responses.</p><p>Wearable devices further allow noninvasive and real-time collection of various vital signs and health-related data, including heart rate, blood pressure, and other biomarkers. Considering these data in MIPD systems holds promise for precision dosing, not only relying on PK parameters (e.g., trough concentrations or area under the plasma drug concentration-time curve results, AUC), but also leveraging novel PD biomarkers and clinical- or patient-related treatment outcomes. Quantitative systems pharmacology and disease modeling can provide a mechanistic framework to integrate PD biomarkers that predict disease progression, treatment response, and drug safety.<span><sup>5</sup></span></p><p>Finally, with the improved ability to collect and securely store extensive amounts of data from both clinical trial and real-world settings, artificial intelligence and machine learning (AI-ML) are growing in importance for identifying patterns of covariates associated with certain clinical outcomes, which could ultimately inform optimal dosing strategies for individual patients.<span><sup>1</sup></span></p><p>Recent technological advancements and research efforts have significantly improved the feasibility of precision dosing in highly complex patients. Evolving MIPD approaches now offer a framework to simultaneously integrate several patient-, drug-, and disease-related covariates known to influence drug exposures and responses.</p><p>A MIPD approach with proof-of-concept data that is yet to be applied prospectively in the clinic is physiologically-based pharmacokinetic (PBPK) modeling and simulation. The terms “virtual twins” and “digital twins” have been coined to describe this type of MIPD, which simulates an individual's PK profile based on their physiology and health status. It is especially appealing for drugs with established target exposure ranges and for special, understudied populations of patients (e.g., children, pregnant women, and lactating mothers) in whom dosing recommendations are unavailable, or when population PK models to guide dosing do not exist.</p><p>AI-ML methods are increasingly studied in precision dosing to predict drug exposure, the incidence of adverse drug reactions, efficacy, or other outcomes.<span><sup>6</sup></span> Examples of pioneering work include the utilization of extreme gradient boosting (XGBoost) models to predict exposures to tacrolimus and mycophenolic acid in support of precision dosing of immunosuppressants.<span><sup>7</sup></span> Additionally, hybrid ML-PK-PD methods, merging AI-ML with traditional pharmacometric approaches, have been investigated to support MIPD systems. A recent study demonstrated that simulation-based data augmentation and enrichment of ML model features with PK-PD model predictions enhanced the predictive ability of the ML model in forecasting chemotherapy-induced neutropenia.<span><sup>8</sup></span> Continuous learning and other approaches to refine PK-PD model parameters and continuously update models used for MIPD have been proposed, and AI-ML could facilitate these processes automatically or semi-automatically as new clinical data are collected through electronic health records (EHRs). In addition, reinforcement learning constitutes a promising approach to enhancing MIPD with the capability of continuous learning.</p><p>Complex dosing and monitoring are difficult to implement, and more advanced precision dosing approaches, including pharmacogenomic (PGx)-guided dosing, are often limited to a few specialties, e.g. hematology, oncology, or pediatrics.<span><sup>9</sup></span> To address clinical implementation, major steps forward in developing user-friendly precision dosing solutions have been achieved in recent years. Furthermore, the successful establishment of precision dosing CDS tools encompasses strategic planning, seamless integration into existing workflows, and continuous improvement. Coupled with the adoption of FHIR (Fast Healthcare Interoperability Resources) standards, these systems can be integrated with major EHRs, paving the way for broader adoption. Companies such as InsightRx and DoseMeRx have successfully demonstrated these integration capabilities for MIPD of approved drugs requiring TDM, including various antimicrobials, immunosuppressants, and anticancer drugs. Moreover, the scope of data within EHRs has been broadening to include a variety of relevant biomarkers and genetic information, facilitating PGx-guided dosing.</p><p>Professional societies are increasingly taking the lead on precision dosing guidelines. This sets the precedent that adherence to drug labeling is softened by professional consensus. Vancomycin dosing and TDM guidelines in 2020 marked a significant milestone in this regard, catalyzing several health systems to implement MIPD strategies aimed at achieving targeted AUC metrics.<span><sup>10</sup></span></p><p>Diverse types of information can inform precision dosing, though necessitating efficient and safe integration from different data sources. The importance of data sharing, while adhering to data protection and privacy regulations, and sometimes with competing commercial interests (e.g., pathology providers), is not to be underestimated, and must be solved for more complex precision dosing approaches to succeed.</p><p>Once precision dosing has been implemented in practice, clinical trials and real-world evidence investigating its benefits over standard approaches are crucial to increase acceptance and foster more widespread adoption. Challenges of conducting such studies include the selection of appropriate precision dosing targets, appropriate end points (e.g., clinical- and patient-reported outcomes), and timing.</p><p>Precision dosing requires additional ethical and legal considerations, including consent from patients and legal protection for prescribers when doses different from those approved are used in the spirit of beneficence. This would be facilitated when guidelines are available from professional societies (Challenge 5).</p><p>Finally, novel reimbursement models for drug therapies are required to encourage precision dosing in drug development and clinical practice. For example, therapy prices independent of the prescribed dose, including outcome-based reimbursement, would accelerate the adoption of precision dosing.</p><p>This perspective emphasizes recent advances in precision dosing and outlines some outstanding opportunities. As precision dosing evolves, greater integration into drug development and clinical practice holds promise to significantly advance personalized medicine, offering more tailored, effective, and safe drug treatments for more patients.</p><p>No funding was received for this work.</p><p>IKM, TM, and TMP declare no conflicts of interest. RWP holds stock in and receives compensation from F Hoffmann la Roche. SG is employed by InsightRx, which produces model-informed precision dosing clinical decision support tools. As an Associate Editor for Clinical Pharmacology &amp; Therapeutics, Iris Minichmayr was not involved in the review or decision process for this paper.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.3000,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cpt.3365","citationCount":"0","resultStr":"{\"title\":\"Recent Advances Addressing the Challenges of Precision Dosing\",\"authors\":\"Iris K. Minichmayr,&nbsp;Tomoyuki Mizuno,&nbsp;Srijib Goswami,&nbsp;Richard W. Peck,&nbsp;Thomas M. Polasek,&nbsp;the American Society of Clinical Pharmacology and Therapeutics Precision Dosing Community\",\"doi\":\"10.1002/cpt.3365\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>Precision dosing can improve drug therapy in complex, critically ill, and chronically ill patient populations who exhibit vast interindividual variabilities in exposures and responses. This perspective outlines five major challenges in precision dosing and highlights their recent progress: (1) application in drug development, (2) improved clinical trials, (3) usefulness of response biomarkers, (4) confidence in model-informed precision dosing, and (5) receptiveness in clinical practice. Many outstanding opportunities in precision dosing remain within regulatory frameworks, data integration and protection, ethics, and reimbursement strategies.</p><p>Precision dosing is a foundational pillar of personalized medicine. The term has been applied to different dosing strategies aimed at improving outcomes and safety in complex patients displaying substantial interindividual variability in drug exposure and response. Depending on their degree of individualization, various “precision dosing methods” result in dosing recommendations for patient subgroups based on specific indications, genotypes, or patient characteristics like renal function, and/or in more individualized dosing based on measurements obtained during drug treatment, for example, drug concentrations from therapeutic drug monitoring (TDM). In complex situations in which target effect measurement and data interpretation are not straightforward, multiple pieces rather than a single pieces of information are ideally combined for precision dosing, requiring sophisticated predictive models.</p><p>Over the past 5–10 years, precision dosing activities, ranging from dried blood spot analyses of plasma drug concentrations to novel models and educational initiatives fostering better collaboration, have surged, with the goals of benefiting patients, aiding prescribers, saving money, and supporting the development of difficult-to-use drugs. This perspective describes recent advances that address five major challenges of precision dosing (<b>Figure</b> 1), followed by a summary of other challenges with slow progress.</p><p>There are strong economic and logistical reasons to limit the number of test doses studied in clinical development. Despite these challenges, precision dosing may be invaluable in tackling complex drug development scenarios, such as for (1) drugs with narrow therapeutic windows, (2) drugs that can lead to severe adverse effects at otherwise typical doses, (3) drugs used for conditions like cancer that carry serious consequences if undertreated, (4) drugs for treating rare, severe, and progressive diseases, and (5) drugs requiring invasive administration routes, for example, intravitreal or intrathecal.<span><sup>1</sup></span></p><p>Regulatory support is variable, but a successful example of pharma-regulatory collaboration supporting precision dosing is MyPKFit®, a clinical decision support (CDS) tool approved alongside recombinant factor VIII that personalizes dosing in hemophilia A. Also, the ophthalmologic drug faricimab was developed with a precision dosing arm in all four of its pivotal clinical trials, covering two indications, that is, neovascular age-related macular degeneration and diabetic macular edema. Personalized treatment with faricimab was approved, with dosing interval individualization according to disease activity such as visual outcomes.<span><sup>1</sup></span></p><p>Generating clinical trial evidence for precision dosing poses a significant challenge with much progress in recent years. As confidence in dose predictions increases (Challenge 4), incorporating MIPD into prospective clinical trials, such as concentration-controlled or effect-controlled trials, is becoming more common.</p><p>Phase II and III studies with individualized dosing arms rather than fixed dosing arms facilitate the exploration of wider and more nuanced doses, resulting in the approval of dose ranges, and subsequently reduced necessity for off-label dosing. Clinical trials of GLP-1 agonists use this approach because there is a fine balance between efficacy and gastrointestinal intolerance.<span><sup>2</sup></span></p><p>In 2021, the FDA's Oncology Center of Excellence initiated Project Optimus to reform the dose selection paradigm in oncology drug development. Project Optimus's primary goal is to ensure that new cancer drugs are developed with optimal dosing strategies by leveraging modern clinical trial designs and more adaptable, patient-centered approaches to dosing – instead of the conventional maximum tolerated dose approach – that consider individual variability and the unique characteristics of the respective cancer type.</p><p>Substantial portions of pharmacokinetic-pharmacodynamic (PK-PD) variability remain unknown for many drugs. Efforts to further understand disease heterogeneity and variability in drug responses, even among patients with comparable drug exposures, have significantly increased in the last decade with the discovery of novel biomarkers and the development of new, more sensitive analysis methods.</p><p>For certain drugs, TDM utilizing microsampling devices – allowing dried blood spot collection at home and delivery to a laboratory for concentration testing – is feasible.<span><sup>3</sup></span> Furthermore, recent advances in biosensor technology have improved the ability to measure drug concentrations directly and non-invasively.<span><sup>4</sup></span> This extends to biomarker monitoring, exemplified by the boom in continuous blood glucose monitoring patches that provide feedback on antidiabetic drug responses.</p><p>Wearable devices further allow noninvasive and real-time collection of various vital signs and health-related data, including heart rate, blood pressure, and other biomarkers. Considering these data in MIPD systems holds promise for precision dosing, not only relying on PK parameters (e.g., trough concentrations or area under the plasma drug concentration-time curve results, AUC), but also leveraging novel PD biomarkers and clinical- or patient-related treatment outcomes. Quantitative systems pharmacology and disease modeling can provide a mechanistic framework to integrate PD biomarkers that predict disease progression, treatment response, and drug safety.<span><sup>5</sup></span></p><p>Finally, with the improved ability to collect and securely store extensive amounts of data from both clinical trial and real-world settings, artificial intelligence and machine learning (AI-ML) are growing in importance for identifying patterns of covariates associated with certain clinical outcomes, which could ultimately inform optimal dosing strategies for individual patients.<span><sup>1</sup></span></p><p>Recent technological advancements and research efforts have significantly improved the feasibility of precision dosing in highly complex patients. Evolving MIPD approaches now offer a framework to simultaneously integrate several patient-, drug-, and disease-related covariates known to influence drug exposures and responses.</p><p>A MIPD approach with proof-of-concept data that is yet to be applied prospectively in the clinic is physiologically-based pharmacokinetic (PBPK) modeling and simulation. The terms “virtual twins” and “digital twins” have been coined to describe this type of MIPD, which simulates an individual's PK profile based on their physiology and health status. It is especially appealing for drugs with established target exposure ranges and for special, understudied populations of patients (e.g., children, pregnant women, and lactating mothers) in whom dosing recommendations are unavailable, or when population PK models to guide dosing do not exist.</p><p>AI-ML methods are increasingly studied in precision dosing to predict drug exposure, the incidence of adverse drug reactions, efficacy, or other outcomes.<span><sup>6</sup></span> Examples of pioneering work include the utilization of extreme gradient boosting (XGBoost) models to predict exposures to tacrolimus and mycophenolic acid in support of precision dosing of immunosuppressants.<span><sup>7</sup></span> Additionally, hybrid ML-PK-PD methods, merging AI-ML with traditional pharmacometric approaches, have been investigated to support MIPD systems. A recent study demonstrated that simulation-based data augmentation and enrichment of ML model features with PK-PD model predictions enhanced the predictive ability of the ML model in forecasting chemotherapy-induced neutropenia.<span><sup>8</sup></span> Continuous learning and other approaches to refine PK-PD model parameters and continuously update models used for MIPD have been proposed, and AI-ML could facilitate these processes automatically or semi-automatically as new clinical data are collected through electronic health records (EHRs). In addition, reinforcement learning constitutes a promising approach to enhancing MIPD with the capability of continuous learning.</p><p>Complex dosing and monitoring are difficult to implement, and more advanced precision dosing approaches, including pharmacogenomic (PGx)-guided dosing, are often limited to a few specialties, e.g. hematology, oncology, or pediatrics.<span><sup>9</sup></span> To address clinical implementation, major steps forward in developing user-friendly precision dosing solutions have been achieved in recent years. Furthermore, the successful establishment of precision dosing CDS tools encompasses strategic planning, seamless integration into existing workflows, and continuous improvement. Coupled with the adoption of FHIR (Fast Healthcare Interoperability Resources) standards, these systems can be integrated with major EHRs, paving the way for broader adoption. Companies such as InsightRx and DoseMeRx have successfully demonstrated these integration capabilities for MIPD of approved drugs requiring TDM, including various antimicrobials, immunosuppressants, and anticancer drugs. Moreover, the scope of data within EHRs has been broadening to include a variety of relevant biomarkers and genetic information, facilitating PGx-guided dosing.</p><p>Professional societies are increasingly taking the lead on precision dosing guidelines. This sets the precedent that adherence to drug labeling is softened by professional consensus. Vancomycin dosing and TDM guidelines in 2020 marked a significant milestone in this regard, catalyzing several health systems to implement MIPD strategies aimed at achieving targeted AUC metrics.<span><sup>10</sup></span></p><p>Diverse types of information can inform precision dosing, though necessitating efficient and safe integration from different data sources. The importance of data sharing, while adhering to data protection and privacy regulations, and sometimes with competing commercial interests (e.g., pathology providers), is not to be underestimated, and must be solved for more complex precision dosing approaches to succeed.</p><p>Once precision dosing has been implemented in practice, clinical trials and real-world evidence investigating its benefits over standard approaches are crucial to increase acceptance and foster more widespread adoption. Challenges of conducting such studies include the selection of appropriate precision dosing targets, appropriate end points (e.g., clinical- and patient-reported outcomes), and timing.</p><p>Precision dosing requires additional ethical and legal considerations, including consent from patients and legal protection for prescribers when doses different from those approved are used in the spirit of beneficence. This would be facilitated when guidelines are available from professional societies (Challenge 5).</p><p>Finally, novel reimbursement models for drug therapies are required to encourage precision dosing in drug development and clinical practice. For example, therapy prices independent of the prescribed dose, including outcome-based reimbursement, would accelerate the adoption of precision dosing.</p><p>This perspective emphasizes recent advances in precision dosing and outlines some outstanding opportunities. As precision dosing evolves, greater integration into drug development and clinical practice holds promise to significantly advance personalized medicine, offering more tailored, effective, and safe drug treatments for more patients.</p><p>No funding was received for this work.</p><p>IKM, TM, and TMP declare no conflicts of interest. RWP holds stock in and receives compensation from F Hoffmann la Roche. SG is employed by InsightRx, which produces model-informed precision dosing clinical decision support tools. As an Associate Editor for Clinical Pharmacology &amp; Therapeutics, Iris Minichmayr was not involved in the review or decision process for this paper.</p>\",\"PeriodicalId\":153,\"journal\":{\"name\":\"Clinical Pharmacology & Therapeutics\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":6.3000,\"publicationDate\":\"2024-08-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cpt.3365\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Clinical Pharmacology & Therapeutics\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/cpt.3365\",\"RegionNum\":2,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"PHARMACOLOGY & PHARMACY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Clinical Pharmacology & Therapeutics","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cpt.3365","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

摘要

3 此外,生物传感器技术的最新进展提高了直接和无创测量药物浓度的能力。4 这也延伸到了生物标志物监测领域,例如可提供抗糖尿病药物反应反馈的连续血糖监测贴片的蓬勃发展。在 MIPD 系统中考虑到这些数据,不仅可以依靠 PK 参数(如谷浓度或血浆药物浓度-时间曲线下面积结果,AUC),还可以利用新型 PD 生物标记物和临床或患者相关的治疗结果,为精确给药带来希望。定量系统药理学和疾病建模可提供一个机理框架,用于整合预测疾病进展、治疗反应和药物安全性的 PD 生物标记物。最后,随着从临床试验和真实世界环境中收集和安全存储大量数据的能力不断提高,人工智能和机器学习(AI-ML)在识别与某些临床结果相关的协变量模式方面的重要性日益凸显,最终可为个体患者的最佳用药策略提供依据。目前,不断发展的 MIPD 方法提供了一个框架,可同时整合已知会影响药物暴露和反应的多个患者、药物和疾病相关协变量。人们创造了 "虚拟双胞胎 "和 "数字双胞胎 "这两个术语来描述这种类型的 MIPD。对于目标暴露范围已经确定的药物和研究不足的特殊患者群体(如儿童、孕妇和哺乳期母亲)来说,这种方法尤其具有吸引力,因为在这些患者群体中还没有用药建议,或者还不存在指导用药的群体 PK 模型。开创性工作的例子包括利用极端梯度提升(XGBoost)模型预测他克莫司和霉酚酸的暴露量,以支持免疫抑制剂的精确用药。最近的一项研究表明,基于模拟的数据增强和 PK-PD 模型预测丰富了 ML 模型的特征,增强了 ML 模型对化疗引起的中性粒细胞减少症的预测能力。此外,强化学习也是增强 MIPD 持续学习能力的一种有前途的方法。复杂的用药和监测很难实施,更先进的精确用药方法,包括药物基因组学(PGx)指导用药,往往仅限于少数专科,如血液科、肿瘤科或儿科。此外,精准给药 CDS 工具的成功建立还包括战略规划、与现有工作流程的无缝整合以及持续改进。再加上 FHIR(快速医疗互操作性资源)标准的采用,这些系统可以与主要的电子病历集成,为更广泛的应用铺平了道路。InsightRx 和 DoseMeRx 等公司已经成功展示了这些集成功能,可用于需要 TDM 的已批准药物的 MIPD,包括各种抗菌药、免疫抑制剂和抗癌药。此外,电子病历中的数据范围也在不断扩大,包括各种相关的生物标记物和遗传信息,为 PGx 指导用药提供了便利。这开创了一个先例,即通过专业共识来软化对药物标签的遵守。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

Recent Advances Addressing the Challenges of Precision Dosing

Recent Advances Addressing the Challenges of Precision Dosing

Precision dosing can improve drug therapy in complex, critically ill, and chronically ill patient populations who exhibit vast interindividual variabilities in exposures and responses. This perspective outlines five major challenges in precision dosing and highlights their recent progress: (1) application in drug development, (2) improved clinical trials, (3) usefulness of response biomarkers, (4) confidence in model-informed precision dosing, and (5) receptiveness in clinical practice. Many outstanding opportunities in precision dosing remain within regulatory frameworks, data integration and protection, ethics, and reimbursement strategies.

Precision dosing is a foundational pillar of personalized medicine. The term has been applied to different dosing strategies aimed at improving outcomes and safety in complex patients displaying substantial interindividual variability in drug exposure and response. Depending on their degree of individualization, various “precision dosing methods” result in dosing recommendations for patient subgroups based on specific indications, genotypes, or patient characteristics like renal function, and/or in more individualized dosing based on measurements obtained during drug treatment, for example, drug concentrations from therapeutic drug monitoring (TDM). In complex situations in which target effect measurement and data interpretation are not straightforward, multiple pieces rather than a single pieces of information are ideally combined for precision dosing, requiring sophisticated predictive models.

Over the past 5–10 years, precision dosing activities, ranging from dried blood spot analyses of plasma drug concentrations to novel models and educational initiatives fostering better collaboration, have surged, with the goals of benefiting patients, aiding prescribers, saving money, and supporting the development of difficult-to-use drugs. This perspective describes recent advances that address five major challenges of precision dosing (Figure 1), followed by a summary of other challenges with slow progress.

There are strong economic and logistical reasons to limit the number of test doses studied in clinical development. Despite these challenges, precision dosing may be invaluable in tackling complex drug development scenarios, such as for (1) drugs with narrow therapeutic windows, (2) drugs that can lead to severe adverse effects at otherwise typical doses, (3) drugs used for conditions like cancer that carry serious consequences if undertreated, (4) drugs for treating rare, severe, and progressive diseases, and (5) drugs requiring invasive administration routes, for example, intravitreal or intrathecal.1

Regulatory support is variable, but a successful example of pharma-regulatory collaboration supporting precision dosing is MyPKFit®, a clinical decision support (CDS) tool approved alongside recombinant factor VIII that personalizes dosing in hemophilia A. Also, the ophthalmologic drug faricimab was developed with a precision dosing arm in all four of its pivotal clinical trials, covering two indications, that is, neovascular age-related macular degeneration and diabetic macular edema. Personalized treatment with faricimab was approved, with dosing interval individualization according to disease activity such as visual outcomes.1

Generating clinical trial evidence for precision dosing poses a significant challenge with much progress in recent years. As confidence in dose predictions increases (Challenge 4), incorporating MIPD into prospective clinical trials, such as concentration-controlled or effect-controlled trials, is becoming more common.

Phase II and III studies with individualized dosing arms rather than fixed dosing arms facilitate the exploration of wider and more nuanced doses, resulting in the approval of dose ranges, and subsequently reduced necessity for off-label dosing. Clinical trials of GLP-1 agonists use this approach because there is a fine balance between efficacy and gastrointestinal intolerance.2

In 2021, the FDA's Oncology Center of Excellence initiated Project Optimus to reform the dose selection paradigm in oncology drug development. Project Optimus's primary goal is to ensure that new cancer drugs are developed with optimal dosing strategies by leveraging modern clinical trial designs and more adaptable, patient-centered approaches to dosing – instead of the conventional maximum tolerated dose approach – that consider individual variability and the unique characteristics of the respective cancer type.

Substantial portions of pharmacokinetic-pharmacodynamic (PK-PD) variability remain unknown for many drugs. Efforts to further understand disease heterogeneity and variability in drug responses, even among patients with comparable drug exposures, have significantly increased in the last decade with the discovery of novel biomarkers and the development of new, more sensitive analysis methods.

For certain drugs, TDM utilizing microsampling devices – allowing dried blood spot collection at home and delivery to a laboratory for concentration testing – is feasible.3 Furthermore, recent advances in biosensor technology have improved the ability to measure drug concentrations directly and non-invasively.4 This extends to biomarker monitoring, exemplified by the boom in continuous blood glucose monitoring patches that provide feedback on antidiabetic drug responses.

Wearable devices further allow noninvasive and real-time collection of various vital signs and health-related data, including heart rate, blood pressure, and other biomarkers. Considering these data in MIPD systems holds promise for precision dosing, not only relying on PK parameters (e.g., trough concentrations or area under the plasma drug concentration-time curve results, AUC), but also leveraging novel PD biomarkers and clinical- or patient-related treatment outcomes. Quantitative systems pharmacology and disease modeling can provide a mechanistic framework to integrate PD biomarkers that predict disease progression, treatment response, and drug safety.5

Finally, with the improved ability to collect and securely store extensive amounts of data from both clinical trial and real-world settings, artificial intelligence and machine learning (AI-ML) are growing in importance for identifying patterns of covariates associated with certain clinical outcomes, which could ultimately inform optimal dosing strategies for individual patients.1

Recent technological advancements and research efforts have significantly improved the feasibility of precision dosing in highly complex patients. Evolving MIPD approaches now offer a framework to simultaneously integrate several patient-, drug-, and disease-related covariates known to influence drug exposures and responses.

A MIPD approach with proof-of-concept data that is yet to be applied prospectively in the clinic is physiologically-based pharmacokinetic (PBPK) modeling and simulation. The terms “virtual twins” and “digital twins” have been coined to describe this type of MIPD, which simulates an individual's PK profile based on their physiology and health status. It is especially appealing for drugs with established target exposure ranges and for special, understudied populations of patients (e.g., children, pregnant women, and lactating mothers) in whom dosing recommendations are unavailable, or when population PK models to guide dosing do not exist.

AI-ML methods are increasingly studied in precision dosing to predict drug exposure, the incidence of adverse drug reactions, efficacy, or other outcomes.6 Examples of pioneering work include the utilization of extreme gradient boosting (XGBoost) models to predict exposures to tacrolimus and mycophenolic acid in support of precision dosing of immunosuppressants.7 Additionally, hybrid ML-PK-PD methods, merging AI-ML with traditional pharmacometric approaches, have been investigated to support MIPD systems. A recent study demonstrated that simulation-based data augmentation and enrichment of ML model features with PK-PD model predictions enhanced the predictive ability of the ML model in forecasting chemotherapy-induced neutropenia.8 Continuous learning and other approaches to refine PK-PD model parameters and continuously update models used for MIPD have been proposed, and AI-ML could facilitate these processes automatically or semi-automatically as new clinical data are collected through electronic health records (EHRs). In addition, reinforcement learning constitutes a promising approach to enhancing MIPD with the capability of continuous learning.

Complex dosing and monitoring are difficult to implement, and more advanced precision dosing approaches, including pharmacogenomic (PGx)-guided dosing, are often limited to a few specialties, e.g. hematology, oncology, or pediatrics.9 To address clinical implementation, major steps forward in developing user-friendly precision dosing solutions have been achieved in recent years. Furthermore, the successful establishment of precision dosing CDS tools encompasses strategic planning, seamless integration into existing workflows, and continuous improvement. Coupled with the adoption of FHIR (Fast Healthcare Interoperability Resources) standards, these systems can be integrated with major EHRs, paving the way for broader adoption. Companies such as InsightRx and DoseMeRx have successfully demonstrated these integration capabilities for MIPD of approved drugs requiring TDM, including various antimicrobials, immunosuppressants, and anticancer drugs. Moreover, the scope of data within EHRs has been broadening to include a variety of relevant biomarkers and genetic information, facilitating PGx-guided dosing.

Professional societies are increasingly taking the lead on precision dosing guidelines. This sets the precedent that adherence to drug labeling is softened by professional consensus. Vancomycin dosing and TDM guidelines in 2020 marked a significant milestone in this regard, catalyzing several health systems to implement MIPD strategies aimed at achieving targeted AUC metrics.10

Diverse types of information can inform precision dosing, though necessitating efficient and safe integration from different data sources. The importance of data sharing, while adhering to data protection and privacy regulations, and sometimes with competing commercial interests (e.g., pathology providers), is not to be underestimated, and must be solved for more complex precision dosing approaches to succeed.

Once precision dosing has been implemented in practice, clinical trials and real-world evidence investigating its benefits over standard approaches are crucial to increase acceptance and foster more widespread adoption. Challenges of conducting such studies include the selection of appropriate precision dosing targets, appropriate end points (e.g., clinical- and patient-reported outcomes), and timing.

Precision dosing requires additional ethical and legal considerations, including consent from patients and legal protection for prescribers when doses different from those approved are used in the spirit of beneficence. This would be facilitated when guidelines are available from professional societies (Challenge 5).

Finally, novel reimbursement models for drug therapies are required to encourage precision dosing in drug development and clinical practice. For example, therapy prices independent of the prescribed dose, including outcome-based reimbursement, would accelerate the adoption of precision dosing.

This perspective emphasizes recent advances in precision dosing and outlines some outstanding opportunities. As precision dosing evolves, greater integration into drug development and clinical practice holds promise to significantly advance personalized medicine, offering more tailored, effective, and safe drug treatments for more patients.

No funding was received for this work.

IKM, TM, and TMP declare no conflicts of interest. RWP holds stock in and receives compensation from F Hoffmann la Roche. SG is employed by InsightRx, which produces model-informed precision dosing clinical decision support tools. As an Associate Editor for Clinical Pharmacology & Therapeutics, Iris Minichmayr was not involved in the review or decision process for this paper.

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
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:481959085
Book学术官方微信