思考罕见,从内到外思考:复杂罕见病药物开发的简单问题方法。

IF 2.4 4区 医学 Q3 PHARMACOLOGY & PHARMACY Journal of clinical pharmacology Pub Date : 2022-12-01 DOI:10.1002/jcph.2173
Indranil Bhattacharya, Mariam A Ahmed
{"title":"思考罕见,从内到外思考:复杂罕见病药物开发的简单问题方法。","authors":"Indranil Bhattacharya, Mariam A Ahmed","doi":"10.1002/jcph.2173","DOIUrl":null,"url":null,"abstract":"Rare disease drug development follows the same principles as in common diseases, that is, the need to meet the statutory standards for safety and effectiveness. However, viewing this through the lens of the benefitrisk profile, given the unmet medical need, regulatory bodies may exhibit flexibility in exercising scientific judgment on whether the evidence qualifies to have met the statutory standards. Figure 1 captures the principle toward rare disease drug approval, which is balancing the unmet medical need and feasibility of evidence generation versus the evidence package. As further illustrated in Figure 1, to balance the evidence package against the unmet medical need and the feasibility, sponsors have to strive toward increasing the weightage of what is known, the certainty box, and reducing what is not known, the uncertainty box. As discussed in the articles within this issue, rare diseases possess unique traits, many of which are not essentially mutually exclusive: small sample size, heterogeneity of the affected population, limited understanding of the disease pathophysiology, and natural history.1–9 It is steadily becoming common knowledge how the multiple traits of rare diseases can complicate drug development; currently, there are >9000 serious and life-threatening rare diseases with not a single therapeutic option.1 While common disease drug development programs are derisked through a stepwise approach using phase 1, 2a, 2b, and 2 phase 3 studies, rare disease drug development programs may follow an accelerated development paradigm. For example, the clinical development plan may constitute a phase 1 followed by a single phase 3 study or even a seamless clinical trial that serves as phase 1 through phase 3. This nontraditional development pathway not only leads to limited data packages with inflated uncertainty but also can exponentiate common drug development issues faced by sponsors. From a benefit-risk context, all these traits and their associated challenges ultimately amalgamate and create barriers in confident decisionmaking for both sponsors and regulators. Clinical pharmacologists can positively influence decision making by reducing uncertainties through establishing a link of causality throughout the continuum of drug development.10 This link of causality is hypothesized to allow clinical translation with confidence and establishes consistency of clinical trial observations with biological rationale. To create this link, a quantitative framework needs to be conceptualized early. Clinical pharmacologists need to integrate internal and external data using relevant tools that provide insights into the important paradigm of right patient, right medication, and right dose. In rare diseases, we need to dig deeper. For example, what is known about the sources of heterogeneity in the small sample size? How is short-term response linked to long-term outcomes? How is the long-term outcome defined? What is the effect of the disease and the drug on different components of the shortand long-term outcomes? Can the end point be further optimized? What is the minimum clinically important difference? Are there external data available with which we could supplement the evidence package, and if so, what is the relevance and quality of the data?","PeriodicalId":15536,"journal":{"name":"Journal of clinical pharmacology","volume":null,"pages":null},"PeriodicalIF":2.4000,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Think Rare, Think Inside and Out: Simple Question-Based Approach to Complex Rare Disease Drug Development.\",\"authors\":\"Indranil Bhattacharya, Mariam A Ahmed\",\"doi\":\"10.1002/jcph.2173\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"Rare disease drug development follows the same principles as in common diseases, that is, the need to meet the statutory standards for safety and effectiveness. However, viewing this through the lens of the benefitrisk profile, given the unmet medical need, regulatory bodies may exhibit flexibility in exercising scientific judgment on whether the evidence qualifies to have met the statutory standards. Figure 1 captures the principle toward rare disease drug approval, which is balancing the unmet medical need and feasibility of evidence generation versus the evidence package. As further illustrated in Figure 1, to balance the evidence package against the unmet medical need and the feasibility, sponsors have to strive toward increasing the weightage of what is known, the certainty box, and reducing what is not known, the uncertainty box. As discussed in the articles within this issue, rare diseases possess unique traits, many of which are not essentially mutually exclusive: small sample size, heterogeneity of the affected population, limited understanding of the disease pathophysiology, and natural history.1–9 It is steadily becoming common knowledge how the multiple traits of rare diseases can complicate drug development; currently, there are >9000 serious and life-threatening rare diseases with not a single therapeutic option.1 While common disease drug development programs are derisked through a stepwise approach using phase 1, 2a, 2b, and 2 phase 3 studies, rare disease drug development programs may follow an accelerated development paradigm. For example, the clinical development plan may constitute a phase 1 followed by a single phase 3 study or even a seamless clinical trial that serves as phase 1 through phase 3. This nontraditional development pathway not only leads to limited data packages with inflated uncertainty but also can exponentiate common drug development issues faced by sponsors. From a benefit-risk context, all these traits and their associated challenges ultimately amalgamate and create barriers in confident decisionmaking for both sponsors and regulators. Clinical pharmacologists can positively influence decision making by reducing uncertainties through establishing a link of causality throughout the continuum of drug development.10 This link of causality is hypothesized to allow clinical translation with confidence and establishes consistency of clinical trial observations with biological rationale. To create this link, a quantitative framework needs to be conceptualized early. Clinical pharmacologists need to integrate internal and external data using relevant tools that provide insights into the important paradigm of right patient, right medication, and right dose. In rare diseases, we need to dig deeper. For example, what is known about the sources of heterogeneity in the small sample size? How is short-term response linked to long-term outcomes? How is the long-term outcome defined? What is the effect of the disease and the drug on different components of the shortand long-term outcomes? Can the end point be further optimized? What is the minimum clinically important difference? Are there external data available with which we could supplement the evidence package, and if so, what is the relevance and quality of the data?\",\"PeriodicalId\":15536,\"journal\":{\"name\":\"Journal of clinical pharmacology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":2.4000,\"publicationDate\":\"2022-12-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Journal of clinical pharmacology\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1002/jcph.2173\",\"RegionNum\":4,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q3\",\"JCRName\":\"PHARMACOLOGY & PHARMACY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal of clinical pharmacology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1002/jcph.2173","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"PHARMACOLOGY & PHARMACY","Score":null,"Total":0}
引用次数: 0
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
Think Rare, Think Inside and Out: Simple Question-Based Approach to Complex Rare Disease Drug Development.
Rare disease drug development follows the same principles as in common diseases, that is, the need to meet the statutory standards for safety and effectiveness. However, viewing this through the lens of the benefitrisk profile, given the unmet medical need, regulatory bodies may exhibit flexibility in exercising scientific judgment on whether the evidence qualifies to have met the statutory standards. Figure 1 captures the principle toward rare disease drug approval, which is balancing the unmet medical need and feasibility of evidence generation versus the evidence package. As further illustrated in Figure 1, to balance the evidence package against the unmet medical need and the feasibility, sponsors have to strive toward increasing the weightage of what is known, the certainty box, and reducing what is not known, the uncertainty box. As discussed in the articles within this issue, rare diseases possess unique traits, many of which are not essentially mutually exclusive: small sample size, heterogeneity of the affected population, limited understanding of the disease pathophysiology, and natural history.1–9 It is steadily becoming common knowledge how the multiple traits of rare diseases can complicate drug development; currently, there are >9000 serious and life-threatening rare diseases with not a single therapeutic option.1 While common disease drug development programs are derisked through a stepwise approach using phase 1, 2a, 2b, and 2 phase 3 studies, rare disease drug development programs may follow an accelerated development paradigm. For example, the clinical development plan may constitute a phase 1 followed by a single phase 3 study or even a seamless clinical trial that serves as phase 1 through phase 3. This nontraditional development pathway not only leads to limited data packages with inflated uncertainty but also can exponentiate common drug development issues faced by sponsors. From a benefit-risk context, all these traits and their associated challenges ultimately amalgamate and create barriers in confident decisionmaking for both sponsors and regulators. Clinical pharmacologists can positively influence decision making by reducing uncertainties through establishing a link of causality throughout the continuum of drug development.10 This link of causality is hypothesized to allow clinical translation with confidence and establishes consistency of clinical trial observations with biological rationale. To create this link, a quantitative framework needs to be conceptualized early. Clinical pharmacologists need to integrate internal and external data using relevant tools that provide insights into the important paradigm of right patient, right medication, and right dose. In rare diseases, we need to dig deeper. For example, what is known about the sources of heterogeneity in the small sample size? How is short-term response linked to long-term outcomes? How is the long-term outcome defined? What is the effect of the disease and the drug on different components of the shortand long-term outcomes? Can the end point be further optimized? What is the minimum clinically important difference? Are there external data available with which we could supplement the evidence package, and if so, what is the relevance and quality of the data?
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
5.10
自引率
3.40%
发文量
176
审稿时长
2 months
期刊介绍: The Journal of Clinical Pharmacology (JCP) is a Human Pharmacology journal designed to provide physicians, pharmacists, research scientists, regulatory scientists, drug developers and academic colleagues a forum to present research in all aspects of Clinical Pharmacology. This includes original research in pharmacokinetics, pharmacogenetics/pharmacogenomics, pharmacometrics, physiologic based pharmacokinetic modeling, drug interactions, therapeutic drug monitoring, regulatory sciences (including unique methods of data analysis), special population studies, drug development, pharmacovigilance, womens’ health, pediatric pharmacology, and pharmacodynamics. Additionally, JCP publishes review articles, commentaries and educational manuscripts. The Journal also serves as an instrument to disseminate Public Policy statements from the American College of Clinical Pharmacology.
期刊最新文献
Natural History and Real-World Data in Rare Diseases: Applications, Limitations, and Future Perspectives. Overview of Clinical Pharmacology Packages of New Drug Applications Approved for the Treatment of Rare Diseases. Regulatory Framework for Drug Development in Rare Diseases. Bridging the Gap With Clinical Pharmacology in Innovative Rare Disease Treatment Modalities: Targeting DNA to RNA to Protein. Think Rare, Think Inside and Out: Simple Question-Based Approach to Complex Rare Disease Drug Development.
×
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