Therapeutic Orphan No More: Role for Clinical Pharmacology and Translational Science in Developing Therapeutics for Rare and Neglected Diseases

IF 6.3 2区 医学 Q1 PHARMACOLOGY & PHARMACY Clinical Pharmacology & Therapeutics Pub Date : 2024-11-14 DOI:10.1002/cpt.3474
Anuradha Ramamoorthy, Islam Younis, Ya-Feng Wen, Mai Mehanna, Kathleen M. Giacomini, Piet H. van der Graaf
{"title":"Therapeutic Orphan No More: Role for Clinical Pharmacology and Translational Science in Developing Therapeutics for Rare and Neglected Diseases","authors":"Anuradha Ramamoorthy,&nbsp;Islam Younis,&nbsp;Ya-Feng Wen,&nbsp;Mai Mehanna,&nbsp;Kathleen M. Giacomini,&nbsp;Piet H. van der Graaf","doi":"10.1002/cpt.3474","DOIUrl":null,"url":null,"abstract":"<p>Rare disease (a disease that affects fewer than 1 in about 2,000 people<span><sup>1</sup></span>) and neglected disease (an infectious disease that is present in lower income countries) have very limited therapeutic options and can be considered as “therapeutic orphans.” Collectively, rare and neglected diseases are estimated to affect nearly 2 billion people worldwide – about 300 million people are estimated to be affected by rare diseases and 1.7 billion by neglected tropical diseases.<span><sup>2, 3</sup></span> However, it is reported that only &lt;5% of rare diseases and a limited number of neglected diseases have therapeutic options available.<span><sup>4, 5</sup></span> That is, even though one in four people globally are affected by these diseases, very limited therapeutic options exist for these patients. In this special issue, we focus on rare diseases to highlight challenges and opportunities in developing therapies for these diseases and showcase the current and future role of clinical pharmacology and translational sciences in addressing the challenges and taking advantage of the opportunities. Additionally, the American Society for Clinical Pharmacology &amp; Therapeutics (ASCPT) 2024 Annual Meeting Award and State-of-Art lectures summarized in this issue also highlight the challenges and opportunities in developing effective therapies for the neglected diseases (e.g., tropical diseases including tuberculosis) and neglected populations (e.g., pregnant women or breastfeeding mothers). Our intention for this special issue of <i>Clinical Pharmacology &amp; Therapeutics</i> (<i>CPT</i>) is that we can all collectively learn from these examples and experiences to accelerate drug development for these therapeutic orphans (<b>Figure</b> 1).</p><p>There are a number of challenges in developing therapies for rare diseases. Because each rare disease afflicts only a small number of patients, the market size is relatively limited, and consequently, developing drugs for many of these diseases may not be considered as being commercially attractive. Additionally, most rare diseases are poorly understood because limited longitudinal natural history data are available, and consequently, more information on disease etiology, pathogenesis, genotype-to-phenotype correlation, disease progression, and outcome is needed. Furthermore, the small patient population size can lead to difficulties in enrolling sufficient numbers of patients in clinical trials and in generating adequate data to determine the benefit–risk profile of the drug. Compounding these challenges, there may be a lack of well-established biomarkers or animal models that can be leveraged during drug development. In sum, insufficient commercial interest and inadequate scientific research make it challenging to develop effective therapeutics for rare diseases.</p><p>Recognizing some of these challenges and limitations, several regulatory agencies and public-private partnerships have focused their efforts on rare and orphan diseases. Of note, recently, in the United States, the Food and Drug Administration (FDA) established the Rare Disease Innovation Hub to align regulatory efforts within FDA, identify and enable innovative approaches to treat rare diseases, and better serve the rare disease community.<span><sup>6</sup></span> In 2022, the European Commission established the Rare Disease Moonshot with a goal to build rare disease centered public–private partnerships to aid optimization of translational research ecosystem, modernize clinical trials, and strengthen the infrastructure that can help shorten the path to diagnosis and subsequent treatment.<span><sup>7</sup></span> In this issue, Ollivier et al.<span><sup>8</sup></span> describe the purpose, strategies, and priorities of the Rare Disease Moonshot as well as the need for public-private collaborations to stimulate the integration of high-quality regulatory science into R&amp;D, support the development of large-scale sustainable infrastructures, and develop tools like the regulatory readiness scale. Also in this issue, Yu et al. describe the similarities and differences in postmarketing studies issued by the FDA and European Medicines Agency (EMA) while highlighting the need for harmonization between regulators and collaboration between the regulators and drug developers.<span><sup>9</sup></span> The call for increased collaboration and harmonization between the FDA and the EMA is also echoed in a recent study by the National Academies that was commissioned by the FDA to focus on the processes for evaluating the safety and efficacy of drugs for rare diseases in the United States and the European Union.<span><sup>2</sup></span></p><p>When developing therapies for rare diseases across different regulatory regions, as Larsson<span><sup>10</sup></span> explains in a Mini Review in this issue, definitions matter. The definition of prevalence that deems a disease to be rare is different among different regulatory regions. This means that a disease that is considered rare in one region may not be considered rare in another region. In certain regions, rare disease may receive an orphan designation which then can confer additional incentives to the drug upon its approval; however, what the designation entails (e.g., see rituximab in<span><sup>10</sup></span>) and what the incentives are varies across regions. Another terminology that does not have a consistent definition across regions is the “ultra-rare” disease. Though ultra-rare diseases are a subset of rare diseases that affect an exceptionally small number of individuals, the numbers are not codified in regulations as has been done for rare diseases. In literature, ultra-rare diseases are thought to affect fewer than 1 in 50,000 people.<span><sup>11</sup></span> Because an extremely limited number of patients are affected by the ultra-rare diseases, these diseases present unique challenges during diagnosis, drug development, and treatment. In terms of prevalence, even rarer are the “n-of-1” diseases that are thought to affect a single individual or at most a couple of individuals, and these are typically attributed to unique genetic variations. Though several “bespoke” therapies (e.g., Milasen) are being developed for these diseases, these therapies also face unique challenges during development and regulatory approval.<span><sup>12</sup></span></p><p>In recent years, there has been an uptick in the approval of orphan drugs. In 2023 alone, more than half of all novel drugs and biologics approved by the FDA were intended to prevent, diagnose, or treat rare diseases.<span><sup>13</sup></span> However, for some rare diseases even when multiple drugs have been approved, unmet medical need can still persist. For instance, several drugs for Duchenne Muscular Dystrophy (DMD) with different mechanisms of action (e.g., small molecule anti-inflammatory drugs, oligonucleotide therapeutics, gene therapy) have been approved either based on clinical outcomes or via the accelerated approval pathway based on dystrophin biomarker changes. In this issue, Konieczny describes the significant unmet medical need that still exists for DMD patients, given the disease's broad range of phenotypes and symptoms.<span><sup>14</sup></span> Similarly for other rare diseases, Ullman et al.<span><sup>15</sup></span> present data on a novel investigational agent with a mechanism of action (substrate reduction therapy) different from an already approved drug (enzyme replacement therapy) to address the unmet need in treating Pompe disease and Huang et al.<span><sup>16</sup></span> review the progress in developing therapies for Huntington's disease by analyzing studies included in ClinicalTrials.gov. However, for many rare diseases, there are still no approved drugs. Using Shwachman–Diamond Syndrome as an example, a disease with no approved therapy, Hars and McReynolds<span><sup>17</sup></span> share an approach for rare disease drug development that may be applicable for other rare diseases as well. They describe the need to develop non-clinical models, to develop/have dedicated ICD-10 code, to conduct longitudinal natural history studies (that can lead to a good understanding of the genetic etiology as well as clinical manifestation of the disease), to collect patient-reported data, and importantly, to engage the patient community, researchers, and regulators to de-risk drug development.</p><p>Advances in genetic analysis have aided in rare disease diagnosis and drug development. In a Mini Review, Yee et al.<span><sup>18</sup></span> describe the consequences of genetic mutations in vitamin B transporters expressed in the blood–brain barrier that result in rare neurologic diseases, and they highlight the potential development of pharmacologic treatments for these diseases. DeLeeuw et al.<span><sup>19</sup></span> describe how emerging modalities, enhanced by the principles of translational science and clinical pharmacology, are addressing challenges associated with rare diseases using SLC6A1 pathogenic variants as an example. Hamdan et al.<span><sup>20</sup></span> use the Autosomal Recessive Cerebellar Ataxias (ARCAs) registry to point to how these data can facilitate planning future trials.</p><p>Registries and real-world data can be instrumental for rare disease drug development by contributing to our understanding of the disease. In a Mini Review, Weberpals and Wang<span><sup>21</sup></span> summarize the contemporary methodological approaches for the comparison of external control arm analysis in rare diseases and outline some of the criticisms and paths forward based on learnings from emulation challenges of randomized trials using real-world data.</p><p>Given the need to embrace various data sources to develop a totality-of-evidence approach, clinical pharmacology is in a unique position to integrate different pieces of information coming from various non-clinical, clinical, and in silico sources. As Ryder posits in this issue, clinical pharmacology is an integrative discipline that can bring together our understanding of disease, pharmacology, response, and response variability to support the development of drugs for rare and ultra-rare diseases.<span><sup>22</sup></span> There is also a vital role for clinical pharmacology in getting the dosage right, particularly in the era of precision medicines as discussed in a recent <i>CPT</i> special issue.<span><sup>23</sup></span> While Project Optimus aims to reform the dose optimization and dose selection paradigm for cancer therapeutics, getting the dosage right is also critically important for both oncological and non-oncological rare diseases. In this issue, Ahmed et al.<span><sup>24</sup></span> describe some of the challenges and opportunities in selecting appropriate doses for novel and emerging modalities, including enzyme replacement therapies, cell and gene therapies, and oligonucleotide therapeutics. They discuss how data from various sources including animal models, ex vivo and in vitro pharmacology studies, model-based predictions, and real-world data can be integrated to select appropriate doses.</p><p>Particularly, modeling and simulation approaches can help overcome certain challenges seen in rare disease drug development as these exposure-based, biological, and pharmaco-statistical models can combine data from preclinical and clinical sources. Mitra et al.<span><sup>25</sup></span> discuss the application of novel quantitative approaches such as quantitative systems pharmacology (QSP), disease progression modeling, artificial intelligence (AI), machine learning (ML), and clinical trial simulations in accelerating drug development for rare diseases. Focusing particularly on QSP, Neves-Zaph and Kaddi<span><sup>26</sup></span> discuss the current state of the art in the application of QSP for rare disease drug development, including generating virtual digital twins of actual patients, simulating mechanistically the disease progression of rare diseases, and accounting for the phenotypic heterogeneity. They describe how QSP virtual populations can be developed to be more diverse than those often enrolled in clinical trials for rare diseases (representing mild, moderate, and severe phenotypes) and support prediction of therapeutic effects in underrepresented patient subpopulations. Additionally, Hamdan et al.<span><sup>20</sup></span> show how analyses of natural history data from ARCA registry can better inform trial designs by allowing for more effective characterization of disease progression.</p><p>Adding to the complexity, nearly half the rare diseases are pediatric rare diseases <span><sup>27</sup></span>. To enroll children in clinical trials, the general expectation is that the potential treatment should provide a prospect of direct benefit. This unique setting (i.e., children with rare diseases) underscores the importance of leveraging clinical pharmacology principles and tools at our disposal. We can integrate knowledge of disease similarity between children and adults (when available) as well as physiological differences between the two populations to evaluate the safety and effectiveness of the investigational drug in children with rare diseases. In this issue, Krishna et al.<span><sup>28</sup></span> summarize the pharmaceutical industry experience in developing drugs for pediatric rare diseases focused on biomarkers and surrogate endpoints, statistical and development considerations, modeling and simulation, and public–private partnerships. They also highlight opportunities related to using master protocols, end-to-end quantitative integration, and sharing of trial level placebo data to understand disease progression. Readers are referred to this White Paper and the one from Ahmed et al.<span><sup>24</sup></span> for further information on challenges and opportunities in the development of therapeutics for pediatric rare diseases.</p><p>Apart from rare diseases, neglected diseases and neglected populations often become therapeutic orphans. In a Perspective in this issue, Attipoe describes the role and contributions of Drugs for Neglected Diseases initiative (DNDi), a non-profit organization that has focused on developing new treatments for neglected diseases focused on patients in Africa, Asia, and Latin America.<span><sup>29</sup></span> In a Mini Review, Waitt et al.<span><sup>30</sup></span> expand on the 2024 Dolores Shockley Award lecture to discuss the ethical imperative to include women of child-bearing potential, pregnant, and breastfeeding individuals in clinical trials including pharmacokinetic studies, if there is an expectation that this patient subpopulation will use the drug. They highlight the importance of community engagement and capacity building within local communities. Mulubwa and Chibale<span><sup>31</sup></span> highlight the need to build models while considering populations not typically enrolled in biomedical research in order to design doses tailored to that specific population. They describe how leveraging in vitro liver subcellular fraction data from Africans, incorporating information on African genetic variants (including considering the frequency of AI-predicted African prevalent drug-metabolizing gene variants to account for pharmacogenetic diversity across the continent), and including in vitro Mtb drug metabolism kinetic data can be used to promote personalization of therapeutics for tuberculosis, a neglected disease with great unmet medical need.</p><p>Patients with rare and neglected diseases often face a long, difficult, and expensive journey to diagnosis and subsequent treatment. As patient and patient advocates share in this issue, it takes a village to develop drugs for these diseases.<span><sup>17, 19</sup></span> Academic institutions, biopharmaceutical companies, patients, patient advocacy groups, regulatory agencies, and healthcare providers will all need to join forces in advancing the treatment of these therapeutic orphans (Figure 1). Addressing the challenges of rare and neglected diseases requires a united effort across multiple disciplines. We call upon the clinical pharmacology and translational science community to collaborate across sectors and disciplines so that we can collectively transform the landscape of rare and neglected diseases, offering hope to millions of patients worldwide.</p><p>No funding was received for this work.</p><p>Islam Younis is an employee of Merck and Co. Inc may own stocks in Merck and Co. Inc. Ya-Feng Wen is an employee of Gilead Sciences and may own stocks in Gilead Sciences. All other authors declare no competing interests for this work.</p><p>Anuradha Ramamoorthy is currently serving on the ASCPT Board of Directors. This article reflects the views of the author and should not be construed to represent the views of FDA.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":"116 6","pages":"1363-1368"},"PeriodicalIF":6.3000,"publicationDate":"2024-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cpt.3474","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Clinical Pharmacology & Therapeutics","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cpt.3474","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

Rare disease (a disease that affects fewer than 1 in about 2,000 people1) and neglected disease (an infectious disease that is present in lower income countries) have very limited therapeutic options and can be considered as “therapeutic orphans.” Collectively, rare and neglected diseases are estimated to affect nearly 2 billion people worldwide – about 300 million people are estimated to be affected by rare diseases and 1.7 billion by neglected tropical diseases.2, 3 However, it is reported that only <5% of rare diseases and a limited number of neglected diseases have therapeutic options available.4, 5 That is, even though one in four people globally are affected by these diseases, very limited therapeutic options exist for these patients. In this special issue, we focus on rare diseases to highlight challenges and opportunities in developing therapies for these diseases and showcase the current and future role of clinical pharmacology and translational sciences in addressing the challenges and taking advantage of the opportunities. Additionally, the American Society for Clinical Pharmacology & Therapeutics (ASCPT) 2024 Annual Meeting Award and State-of-Art lectures summarized in this issue also highlight the challenges and opportunities in developing effective therapies for the neglected diseases (e.g., tropical diseases including tuberculosis) and neglected populations (e.g., pregnant women or breastfeeding mothers). Our intention for this special issue of Clinical Pharmacology & Therapeutics (CPT) is that we can all collectively learn from these examples and experiences to accelerate drug development for these therapeutic orphans (Figure 1).

There are a number of challenges in developing therapies for rare diseases. Because each rare disease afflicts only a small number of patients, the market size is relatively limited, and consequently, developing drugs for many of these diseases may not be considered as being commercially attractive. Additionally, most rare diseases are poorly understood because limited longitudinal natural history data are available, and consequently, more information on disease etiology, pathogenesis, genotype-to-phenotype correlation, disease progression, and outcome is needed. Furthermore, the small patient population size can lead to difficulties in enrolling sufficient numbers of patients in clinical trials and in generating adequate data to determine the benefit–risk profile of the drug. Compounding these challenges, there may be a lack of well-established biomarkers or animal models that can be leveraged during drug development. In sum, insufficient commercial interest and inadequate scientific research make it challenging to develop effective therapeutics for rare diseases.

Recognizing some of these challenges and limitations, several regulatory agencies and public-private partnerships have focused their efforts on rare and orphan diseases. Of note, recently, in the United States, the Food and Drug Administration (FDA) established the Rare Disease Innovation Hub to align regulatory efforts within FDA, identify and enable innovative approaches to treat rare diseases, and better serve the rare disease community.6 In 2022, the European Commission established the Rare Disease Moonshot with a goal to build rare disease centered public–private partnerships to aid optimization of translational research ecosystem, modernize clinical trials, and strengthen the infrastructure that can help shorten the path to diagnosis and subsequent treatment.7 In this issue, Ollivier et al.8 describe the purpose, strategies, and priorities of the Rare Disease Moonshot as well as the need for public-private collaborations to stimulate the integration of high-quality regulatory science into R&D, support the development of large-scale sustainable infrastructures, and develop tools like the regulatory readiness scale. Also in this issue, Yu et al. describe the similarities and differences in postmarketing studies issued by the FDA and European Medicines Agency (EMA) while highlighting the need for harmonization between regulators and collaboration between the regulators and drug developers.9 The call for increased collaboration and harmonization between the FDA and the EMA is also echoed in a recent study by the National Academies that was commissioned by the FDA to focus on the processes for evaluating the safety and efficacy of drugs for rare diseases in the United States and the European Union.2

When developing therapies for rare diseases across different regulatory regions, as Larsson10 explains in a Mini Review in this issue, definitions matter. The definition of prevalence that deems a disease to be rare is different among different regulatory regions. This means that a disease that is considered rare in one region may not be considered rare in another region. In certain regions, rare disease may receive an orphan designation which then can confer additional incentives to the drug upon its approval; however, what the designation entails (e.g., see rituximab in10) and what the incentives are varies across regions. Another terminology that does not have a consistent definition across regions is the “ultra-rare” disease. Though ultra-rare diseases are a subset of rare diseases that affect an exceptionally small number of individuals, the numbers are not codified in regulations as has been done for rare diseases. In literature, ultra-rare diseases are thought to affect fewer than 1 in 50,000 people.11 Because an extremely limited number of patients are affected by the ultra-rare diseases, these diseases present unique challenges during diagnosis, drug development, and treatment. In terms of prevalence, even rarer are the “n-of-1” diseases that are thought to affect a single individual or at most a couple of individuals, and these are typically attributed to unique genetic variations. Though several “bespoke” therapies (e.g., Milasen) are being developed for these diseases, these therapies also face unique challenges during development and regulatory approval.12

In recent years, there has been an uptick in the approval of orphan drugs. In 2023 alone, more than half of all novel drugs and biologics approved by the FDA were intended to prevent, diagnose, or treat rare diseases.13 However, for some rare diseases even when multiple drugs have been approved, unmet medical need can still persist. For instance, several drugs for Duchenne Muscular Dystrophy (DMD) with different mechanisms of action (e.g., small molecule anti-inflammatory drugs, oligonucleotide therapeutics, gene therapy) have been approved either based on clinical outcomes or via the accelerated approval pathway based on dystrophin biomarker changes. In this issue, Konieczny describes the significant unmet medical need that still exists for DMD patients, given the disease's broad range of phenotypes and symptoms.14 Similarly for other rare diseases, Ullman et al.15 present data on a novel investigational agent with a mechanism of action (substrate reduction therapy) different from an already approved drug (enzyme replacement therapy) to address the unmet need in treating Pompe disease and Huang et al.16 review the progress in developing therapies for Huntington's disease by analyzing studies included in ClinicalTrials.gov. However, for many rare diseases, there are still no approved drugs. Using Shwachman–Diamond Syndrome as an example, a disease with no approved therapy, Hars and McReynolds17 share an approach for rare disease drug development that may be applicable for other rare diseases as well. They describe the need to develop non-clinical models, to develop/have dedicated ICD-10 code, to conduct longitudinal natural history studies (that can lead to a good understanding of the genetic etiology as well as clinical manifestation of the disease), to collect patient-reported data, and importantly, to engage the patient community, researchers, and regulators to de-risk drug development.

Advances in genetic analysis have aided in rare disease diagnosis and drug development. In a Mini Review, Yee et al.18 describe the consequences of genetic mutations in vitamin B transporters expressed in the blood–brain barrier that result in rare neurologic diseases, and they highlight the potential development of pharmacologic treatments for these diseases. DeLeeuw et al.19 describe how emerging modalities, enhanced by the principles of translational science and clinical pharmacology, are addressing challenges associated with rare diseases using SLC6A1 pathogenic variants as an example. Hamdan et al.20 use the Autosomal Recessive Cerebellar Ataxias (ARCAs) registry to point to how these data can facilitate planning future trials.

Registries and real-world data can be instrumental for rare disease drug development by contributing to our understanding of the disease. In a Mini Review, Weberpals and Wang21 summarize the contemporary methodological approaches for the comparison of external control arm analysis in rare diseases and outline some of the criticisms and paths forward based on learnings from emulation challenges of randomized trials using real-world data.

Given the need to embrace various data sources to develop a totality-of-evidence approach, clinical pharmacology is in a unique position to integrate different pieces of information coming from various non-clinical, clinical, and in silico sources. As Ryder posits in this issue, clinical pharmacology is an integrative discipline that can bring together our understanding of disease, pharmacology, response, and response variability to support the development of drugs for rare and ultra-rare diseases.22 There is also a vital role for clinical pharmacology in getting the dosage right, particularly in the era of precision medicines as discussed in a recent CPT special issue.23 While Project Optimus aims to reform the dose optimization and dose selection paradigm for cancer therapeutics, getting the dosage right is also critically important for both oncological and non-oncological rare diseases. In this issue, Ahmed et al.24 describe some of the challenges and opportunities in selecting appropriate doses for novel and emerging modalities, including enzyme replacement therapies, cell and gene therapies, and oligonucleotide therapeutics. They discuss how data from various sources including animal models, ex vivo and in vitro pharmacology studies, model-based predictions, and real-world data can be integrated to select appropriate doses.

Particularly, modeling and simulation approaches can help overcome certain challenges seen in rare disease drug development as these exposure-based, biological, and pharmaco-statistical models can combine data from preclinical and clinical sources. Mitra et al.25 discuss the application of novel quantitative approaches such as quantitative systems pharmacology (QSP), disease progression modeling, artificial intelligence (AI), machine learning (ML), and clinical trial simulations in accelerating drug development for rare diseases. Focusing particularly on QSP, Neves-Zaph and Kaddi26 discuss the current state of the art in the application of QSP for rare disease drug development, including generating virtual digital twins of actual patients, simulating mechanistically the disease progression of rare diseases, and accounting for the phenotypic heterogeneity. They describe how QSP virtual populations can be developed to be more diverse than those often enrolled in clinical trials for rare diseases (representing mild, moderate, and severe phenotypes) and support prediction of therapeutic effects in underrepresented patient subpopulations. Additionally, Hamdan et al.20 show how analyses of natural history data from ARCA registry can better inform trial designs by allowing for more effective characterization of disease progression.

Adding to the complexity, nearly half the rare diseases are pediatric rare diseases 27. To enroll children in clinical trials, the general expectation is that the potential treatment should provide a prospect of direct benefit. This unique setting (i.e., children with rare diseases) underscores the importance of leveraging clinical pharmacology principles and tools at our disposal. We can integrate knowledge of disease similarity between children and adults (when available) as well as physiological differences between the two populations to evaluate the safety and effectiveness of the investigational drug in children with rare diseases. In this issue, Krishna et al.28 summarize the pharmaceutical industry experience in developing drugs for pediatric rare diseases focused on biomarkers and surrogate endpoints, statistical and development considerations, modeling and simulation, and public–private partnerships. They also highlight opportunities related to using master protocols, end-to-end quantitative integration, and sharing of trial level placebo data to understand disease progression. Readers are referred to this White Paper and the one from Ahmed et al.24 for further information on challenges and opportunities in the development of therapeutics for pediatric rare diseases.

Apart from rare diseases, neglected diseases and neglected populations often become therapeutic orphans. In a Perspective in this issue, Attipoe describes the role and contributions of Drugs for Neglected Diseases initiative (DNDi), a non-profit organization that has focused on developing new treatments for neglected diseases focused on patients in Africa, Asia, and Latin America.29 In a Mini Review, Waitt et al.30 expand on the 2024 Dolores Shockley Award lecture to discuss the ethical imperative to include women of child-bearing potential, pregnant, and breastfeeding individuals in clinical trials including pharmacokinetic studies, if there is an expectation that this patient subpopulation will use the drug. They highlight the importance of community engagement and capacity building within local communities. Mulubwa and Chibale31 highlight the need to build models while considering populations not typically enrolled in biomedical research in order to design doses tailored to that specific population. They describe how leveraging in vitro liver subcellular fraction data from Africans, incorporating information on African genetic variants (including considering the frequency of AI-predicted African prevalent drug-metabolizing gene variants to account for pharmacogenetic diversity across the continent), and including in vitro Mtb drug metabolism kinetic data can be used to promote personalization of therapeutics for tuberculosis, a neglected disease with great unmet medical need.

Patients with rare and neglected diseases often face a long, difficult, and expensive journey to diagnosis and subsequent treatment. As patient and patient advocates share in this issue, it takes a village to develop drugs for these diseases.17, 19 Academic institutions, biopharmaceutical companies, patients, patient advocacy groups, regulatory agencies, and healthcare providers will all need to join forces in advancing the treatment of these therapeutic orphans (Figure 1). Addressing the challenges of rare and neglected diseases requires a united effort across multiple disciplines. We call upon the clinical pharmacology and translational science community to collaborate across sectors and disciplines so that we can collectively transform the landscape of rare and neglected diseases, offering hope to millions of patients worldwide.

No funding was received for this work.

Islam Younis is an employee of Merck and Co. Inc may own stocks in Merck and Co. Inc. Ya-Feng Wen is an employee of Gilead Sciences and may own stocks in Gilead Sciences. All other authors declare no competing interests for this work.

Anuradha Ramamoorthy is currently serving on the ASCPT Board of Directors. This article reflects the views of the author and should not be construed to represent the views of FDA.

Abstract Image

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
不再是治疗孤儿:临床药理学和转化科学在开发罕见和被忽视疾病治疗药物中的作用。
22 临床药理学在正确掌握剂量方面也发挥着重要作用,尤其是在最近一期 CPT 特刊所讨论的精准药物时代。23 虽然 "Optimus 项目 "旨在改革癌症疗法的剂量优化和剂量选择模式,但正确掌握剂量对于肿瘤和非肿瘤罕见病同样至关重要。在本期杂志中,Ahmed 等人24 描述了为新型和新兴疗法(包括酶替代疗法、细胞和基因疗法以及寡核苷酸疗法)选择适当剂量时所面临的一些挑战和机遇。他们讨论了如何整合各种来源的数据,包括动物模型、体内外药理学研究、基于模型的预测和真实世界数据,以选择合适的剂量。特别是,建模和模拟方法有助于克服罕见病药物开发中的某些挑战,因为这些基于暴露、生物和药物统计的模型可以整合临床前和临床来源的数据。Mitra 等人25 讨论了新型定量方法在加速罕见病药物开发中的应用,如定量系统药理学(QSP)、疾病进展建模、人工智能(AI)、机器学习(ML)和临床试验模拟。Neves-Zaph 和 Kaddi26 特别关注 QSP,讨论了 QSP 在罕见病药物开发中的应用现状,包括生成实际患者的虚拟数字双胞胎、从机理上模拟罕见病的疾病进展以及考虑表型异质性。他们介绍了如何开发 QSP 虚拟人群,使其比罕见病临床试验中经常登记的人群(代表轻度、中度和重度表型)更加多样化,并支持对代表性不足的患者亚群的治疗效果进行预测。此外,Hamdan 等人20 还展示了如何通过分析 ARCA 登记册中的自然病史数据来更有效地描述疾病进展特征,从而更好地为试验设计提供信息。要让儿童参与临床试验,一般的期望是潜在的治疗方法应能提供直接获益的前景。这种独特的环境(即罕见病患儿)凸显了利用临床药理学原理和我们所掌握的工具的重要性。我们可以整合儿童与成人疾病相似性(如有)以及两类人群生理差异的知识,评估研究药物对罕见病儿童的安全性和有效性。在本期中,Krishna 等人28 总结了制药行业在开发儿科罕见病药物方面的经验,重点关注生物标记物和替代终点、统计和开发注意事项、建模和模拟以及公私合作伙伴关系。他们还强调了与使用主协议、端到端定量整合以及共享试验水平安慰剂数据以了解疾病进展相关的机遇。读者可参阅本白皮书和 Ahmed 等人的白皮书24 ,进一步了解儿科罕见病疗法开发中的挑战和机遇。在本期的一篇 "视角 "文章中,Attipoe 介绍了 "被忽视疾病药物倡议"(DNDi)的作用和贡献。在一篇小型评论中,Waitt 等人30 对 2024 年多洛雷斯-肖克利奖的演讲内容进行了扩展,讨论了将育龄妇女、孕妇和哺乳期妇女纳入临床试验(包括药代动力学研究)的伦理必要性,前提是预期该患者亚群将使用该药物。他们强调了当地社区参与和能力建设的重要性。Mulubwa 和 Chibale31 强调有必要在建立模型的同时考虑到通常不参与生物医学研究的人群,以便设计出适合特定人群的剂量。他们介绍了如何利用非洲人的体外肝脏亚细胞分馏数据,纳入非洲基因变异信息(包括考虑人工智能预测的非洲流行药物代谢基因变异频率,以考虑整个非洲大陆的药物遗传多样性),并纳入体外 Mtb 药物代谢动力学数据,以促进结核病治疗方法的个性化,结核病是一种被忽视的疾病,有大量医疗需求未得到满足。 罕见病和被忽视疾病患者往往面临漫长、艰难和昂贵的诊断和治疗过程。17、19 学术机构、生物制药公司、患者、患者权益组织、监管机构和医疗服务提供者都需要联合起来,共同推进这些治疗孤儿的治疗工作(图 1)。应对罕见病和被忽视疾病的挑战需要多个学科的共同努力。我们呼吁临床药理学和转化科学界跨部门、跨学科合作,共同改变罕见病和被忽视疾病的面貌,为全球数百万患者带来希望。Inc 可能拥有默克公司的股票。公司的股票。文雅锋是吉利德科学公司的员工,可能拥有吉利德科学公司的股票。Anuradha Ramamoorthy目前是ASCPT董事会成员。本文仅代表作者个人观点,不应被视为代表 FDA 的观点。
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 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.
期刊最新文献
Artificial Intelligence in European Medicines Regulation: From Vision to Action. Harnessing the Capabilities of Artificial Intelligence for the Benefit of Public and Animal Health. Mechanistic Modeling of Spatial Heterogeneity of Drug Penetration and Exposure in the Human Central Nervous System and Brain Tumors. Pharmacokinetic Modeling and Model-Based Hypothesis Generation for Dose Optimization of Clonidine in Neonates With Neonatal Opioid Withdrawal Syndrome. Planning and Implementing Master Protocol Trials in Japan: Key Considerations of the Japanese Guideline. Cerebral Ischemia Protection After Aneurysmal Subarachnoid Hemorrhage: CSF Nimodipine Levels After Intravenous Versus Oral Nimodipine Administration.
×
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
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1