利用蛋白质组学互换生物类似物。

IF 5.5 2区 医学 Q1 PHARMACOLOGY & PHARMACY
Piet H. van der Graaf, Ingolf Cascorbi
{"title":"利用蛋白质组学互换生物类似物。","authors":"Piet H. van der Graaf,&nbsp;Ingolf Cascorbi","doi":"10.1002/cpt.70032","DOIUrl":null,"url":null,"abstract":"<p>The development and implementation of bioequivalence (BE) approaches can be considered one of the major innovations in clinical pharmacology.<span><sup>1</sup></span> Before the 1984 Waxman-Hatch Act (Drug Price Competition and Patent Term Restoration Act) created the Abbreviated New Drug Application as a regulatory approval path using BE, only 19% of drugs prescribed in the United States (US) were generic. This number has now increased to ~90%, saving society and the healthcare system trillions of dollars and making treatments available to patients across the world who could previously not afford them.<span><sup>1</sup></span></p><p>Because exact molecular copies cannot be made of a biological therapeutic, there is no such thing as a “generic biological.” Even if the amino acid sequence of a therapeutic protein is completely identical, there may be changes in the glycosylation pattern, differences in protein folding or oxidation or deamination, changes at the start or end of the protein (e.g., loss of terminal amino acids through enzymatic processes) or changes in the grade of aggregation. Therefore, demonstrating BE alone is not sufficient to claim biosimilarity. As a result, randomized, controlled non-inferiority or equivalence studies must generally be conducted to demonstrate the efficacy and safety of the biosimilar compared to the original, making the path to approval complex and costly.<span><sup>2</sup></span> The best we can aim to achieve is a “biosimilar”: a biological product that is highly similar to and has no clinically meaningful differences from an approved reference product.<span><sup>3</sup></span></p><p>As a result, despite the significant shift in recent decades from small molecules drugs to biotherapeutics in drug development and clinical practice, most biologicals do not have an approved biosimilar. There is therefore a huge need to make biosimilar development and approval more efficient, streamlined, and cheaper. Achieving this through clinical pharmacology innovations will be a future milestone of major impacts of our discipline and elevate pharmaco-equity in global healthcare.</p><p>The importance of this topic was illustrated by the January 2023 issue of <i>Clinical Pharmacology &amp; Therapeutics</i> (<i>CPT</i>), which was entirely dedicated to the theme of “Advancing Innovations in Biosimilars.”<span><sup>3</sup></span> The paper by Chekka and colleagues from the US Food and Drug Administration (FDA) in the current issue (<b>Figure</b> 1) reports the latest innovation in regulatory science and how proteomic pharmacodynamic (PD) biomarkers can be used in biosimilarity assessment to reduce the need for comparative clinical efficacy studies.<span><sup>4</sup></span> The authors analyzed candidate proteins from longitudinal plasma samples from healthy subjects who received interferon (IFN)β-1a or pegylated (peg)IFNβ-1a. Several hundred candidates were differentially expressed compared to the placebo and nine were prioritized as potential PD biomarkers common to both biologics. Subsequently, the identified biomarkers were further evaluated using FDA's criteria for desirable characteristics for a PD biomarker for biosimilarity assessment, such as a true dose–response relationship, relevance to mechanism of action, return to baseline post-treatment, sensitivity, and low variability. This finally resulted in the selection of three candidates (Lymphocyte activation gene 3 protein [LAG3], granulins [GRN], and CXC motif chemokine 11 [I-TAC]) that may have the greatest potential as PD biomarker for INFβ-1a biosimilars. It should be emphasized that not only is the reproducibility of the results of great importance, but upstream pathway analyses should also be performed to demonstrate the causal relationship between the respective biological drug and the identified biomarkers. This was the case for all three candidates mentioned above.</p><p>Uncertainty in regulatory acceptance has been a main barrier in the utilization of PD biomarkers for biosimilar development.<span><sup>5</sup></span> The study by Chekka <i>et al</i>. is the first study that provides proof-of-concept that proteomics can be used to identify and develop PD biomarkers to demonstrate biosimilarity and provides a framework for how this can be applied across biosimilar development and approval.</p><p>No funding was received for this work.</p><p>The author declared no competing interests for this work.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":"118 4","pages":"757-758"},"PeriodicalIF":5.5000,"publicationDate":"2025-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/cpt.70032","citationCount":"0","resultStr":"{\"title\":\"Interchanging Biosimilars Using Proteomics\",\"authors\":\"Piet H. van der Graaf,&nbsp;Ingolf Cascorbi\",\"doi\":\"10.1002/cpt.70032\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>The development and implementation of bioequivalence (BE) approaches can be considered one of the major innovations in clinical pharmacology.<span><sup>1</sup></span> Before the 1984 Waxman-Hatch Act (Drug Price Competition and Patent Term Restoration Act) created the Abbreviated New Drug Application as a regulatory approval path using BE, only 19% of drugs prescribed in the United States (US) were generic. This number has now increased to ~90%, saving society and the healthcare system trillions of dollars and making treatments available to patients across the world who could previously not afford them.<span><sup>1</sup></span></p><p>Because exact molecular copies cannot be made of a biological therapeutic, there is no such thing as a “generic biological.” Even if the amino acid sequence of a therapeutic protein is completely identical, there may be changes in the glycosylation pattern, differences in protein folding or oxidation or deamination, changes at the start or end of the protein (e.g., loss of terminal amino acids through enzymatic processes) or changes in the grade of aggregation. Therefore, demonstrating BE alone is not sufficient to claim biosimilarity. As a result, randomized, controlled non-inferiority or equivalence studies must generally be conducted to demonstrate the efficacy and safety of the biosimilar compared to the original, making the path to approval complex and costly.<span><sup>2</sup></span> The best we can aim to achieve is a “biosimilar”: a biological product that is highly similar to and has no clinically meaningful differences from an approved reference product.<span><sup>3</sup></span></p><p>As a result, despite the significant shift in recent decades from small molecules drugs to biotherapeutics in drug development and clinical practice, most biologicals do not have an approved biosimilar. There is therefore a huge need to make biosimilar development and approval more efficient, streamlined, and cheaper. Achieving this through clinical pharmacology innovations will be a future milestone of major impacts of our discipline and elevate pharmaco-equity in global healthcare.</p><p>The importance of this topic was illustrated by the January 2023 issue of <i>Clinical Pharmacology &amp; Therapeutics</i> (<i>CPT</i>), which was entirely dedicated to the theme of “Advancing Innovations in Biosimilars.”<span><sup>3</sup></span> The paper by Chekka and colleagues from the US Food and Drug Administration (FDA) in the current issue (<b>Figure</b> 1) reports the latest innovation in regulatory science and how proteomic pharmacodynamic (PD) biomarkers can be used in biosimilarity assessment to reduce the need for comparative clinical efficacy studies.<span><sup>4</sup></span> The authors analyzed candidate proteins from longitudinal plasma samples from healthy subjects who received interferon (IFN)β-1a or pegylated (peg)IFNβ-1a. Several hundred candidates were differentially expressed compared to the placebo and nine were prioritized as potential PD biomarkers common to both biologics. Subsequently, the identified biomarkers were further evaluated using FDA's criteria for desirable characteristics for a PD biomarker for biosimilarity assessment, such as a true dose–response relationship, relevance to mechanism of action, return to baseline post-treatment, sensitivity, and low variability. This finally resulted in the selection of three candidates (Lymphocyte activation gene 3 protein [LAG3], granulins [GRN], and CXC motif chemokine 11 [I-TAC]) that may have the greatest potential as PD biomarker for INFβ-1a biosimilars. It should be emphasized that not only is the reproducibility of the results of great importance, but upstream pathway analyses should also be performed to demonstrate the causal relationship between the respective biological drug and the identified biomarkers. This was the case for all three candidates mentioned above.</p><p>Uncertainty in regulatory acceptance has been a main barrier in the utilization of PD biomarkers for biosimilar development.<span><sup>5</sup></span> The study by Chekka <i>et al</i>. is the first study that provides proof-of-concept that proteomics can be used to identify and develop PD biomarkers to demonstrate biosimilarity and provides a framework for how this can be applied across biosimilar development and approval.</p><p>No funding was received for this work.</p><p>The author declared no competing interests for this work.</p>\",\"PeriodicalId\":153,\"journal\":{\"name\":\"Clinical Pharmacology & Therapeutics\",\"volume\":\"118 4\",\"pages\":\"757-758\"},\"PeriodicalIF\":5.5000,\"publicationDate\":\"2025-09-16\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://ascpt.onlinelibrary.wiley.com/doi/epdf/10.1002/cpt.70032\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Clinical Pharmacology & Therapeutics\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://ascpt.onlinelibrary.wiley.com/doi/10.1002/cpt.70032\",\"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://ascpt.onlinelibrary.wiley.com/doi/10.1002/cpt.70032","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

生物等效性(BE)方法的发展和实施可以被认为是临床药理学的主要创新之一在1984年Waxman-Hatch法案(药品价格竞争和专利期限恢复法案)将简化新药申请作为使用BE的监管批准途径之前,美国处方的药物中只有19%是仿制药。这一数字现已增加到约90%,为社会和医疗保健系统节省了数万亿美元,并为世界各地以前无法负担治疗费用的患者提供了治疗。由于生物治疗药物无法精确地复制分子,因此不存在“通用生物”这种东西。即使治疗性蛋白质的氨基酸序列完全相同,糖基化模式也可能发生变化,蛋白质折叠或氧化或脱胺的差异,蛋白质开始或结束的变化(例如,酶促过程中末端氨基酸的损失)或聚集等级的变化。因此,仅证明BE不足以主张生物相似性。因此,通常必须进行随机对照非劣效性或等效性研究,以证明生物仿制药与原药相比的有效性和安全性,这使得批准之路复杂且成本高昂我们能达到的最佳目标是“生物仿制药”:一种与已批准的参考产品高度相似且没有临床意义差异的生物制品。因此,尽管近几十年来在药物开发和临床实践中从小分子药物到生物治疗药物发生了重大转变,但大多数生物制剂没有获得批准的生物仿制药。因此,迫切需要使生物类似药的开发和批准更高效、更精简、更便宜。通过临床药理学创新实现这一目标将是我们学科重大影响的未来里程碑,并提升全球医疗保健中的药物公平。2023年1月的《临床药理学与治疗学》(CPT)表明了这一主题的重要性,该杂志完全致力于“推进生物仿制药的创新”这一主题。美国食品和药物管理局(FDA)的Chekka及其同事在本期杂志上发表的论文(图1)报道了监管科学的最新创新,以及蛋白质组药效学(PD)生物标志物如何用于生物相似性评估,以减少对比较临床疗效研究的需求作者分析了接受干扰素(IFN)β-1a或聚乙二醇化(peg)IFNβ-1a治疗的健康受试者纵向血浆样本中的候选蛋白。与安慰剂相比,数百种候选物存在差异表达,其中9种被优先考虑为两种生物制剂共同的潜在PD生物标志物。随后,使用FDA的PD生物标志物生物相似性评估标准对鉴定的生物标志物进行进一步评估,如真实的剂量-反应关系、与作用机制的相关性、治疗后恢复基线、敏感性和低可变性。这最终导致了三个候选物(淋巴细胞活化基因3蛋白[LAG3],颗粒蛋白[GRN]和CXC基序趋化因子11 [I-TAC])的选择,它们可能最有潜力作为INFβ-1a生物仿制药的PD生物标志物。应该强调的是,不仅结果的可重复性非常重要,而且上游通路分析也应该进行,以证明各自的生物药物和鉴定的生物标志物之间的因果关系。上述三位候选人都是如此。监管机构接受度的不确定性一直是PD生物标志物用于生物仿制药开发的主要障碍Chekka等人的研究首次提供了概念验证,即蛋白质组学可用于识别和开发PD生物标志物,以证明生物相似性,并为如何将其应用于生物类似药的开发和批准提供了框架。这项工作没有收到任何资金。作者声明对这项工作没有竞争利益。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

Interchanging Biosimilars Using Proteomics

Interchanging Biosimilars Using Proteomics

Interchanging Biosimilars Using Proteomics

Interchanging Biosimilars Using Proteomics

Interchanging Biosimilars Using Proteomics

The development and implementation of bioequivalence (BE) approaches can be considered one of the major innovations in clinical pharmacology.1 Before the 1984 Waxman-Hatch Act (Drug Price Competition and Patent Term Restoration Act) created the Abbreviated New Drug Application as a regulatory approval path using BE, only 19% of drugs prescribed in the United States (US) were generic. This number has now increased to ~90%, saving society and the healthcare system trillions of dollars and making treatments available to patients across the world who could previously not afford them.1

Because exact molecular copies cannot be made of a biological therapeutic, there is no such thing as a “generic biological.” Even if the amino acid sequence of a therapeutic protein is completely identical, there may be changes in the glycosylation pattern, differences in protein folding or oxidation or deamination, changes at the start or end of the protein (e.g., loss of terminal amino acids through enzymatic processes) or changes in the grade of aggregation. Therefore, demonstrating BE alone is not sufficient to claim biosimilarity. As a result, randomized, controlled non-inferiority or equivalence studies must generally be conducted to demonstrate the efficacy and safety of the biosimilar compared to the original, making the path to approval complex and costly.2 The best we can aim to achieve is a “biosimilar”: a biological product that is highly similar to and has no clinically meaningful differences from an approved reference product.3

As a result, despite the significant shift in recent decades from small molecules drugs to biotherapeutics in drug development and clinical practice, most biologicals do not have an approved biosimilar. There is therefore a huge need to make biosimilar development and approval more efficient, streamlined, and cheaper. Achieving this through clinical pharmacology innovations will be a future milestone of major impacts of our discipline and elevate pharmaco-equity in global healthcare.

The importance of this topic was illustrated by the January 2023 issue of Clinical Pharmacology & Therapeutics (CPT), which was entirely dedicated to the theme of “Advancing Innovations in Biosimilars.”3 The paper by Chekka and colleagues from the US Food and Drug Administration (FDA) in the current issue (Figure 1) reports the latest innovation in regulatory science and how proteomic pharmacodynamic (PD) biomarkers can be used in biosimilarity assessment to reduce the need for comparative clinical efficacy studies.4 The authors analyzed candidate proteins from longitudinal plasma samples from healthy subjects who received interferon (IFN)β-1a or pegylated (peg)IFNβ-1a. Several hundred candidates were differentially expressed compared to the placebo and nine were prioritized as potential PD biomarkers common to both biologics. Subsequently, the identified biomarkers were further evaluated using FDA's criteria for desirable characteristics for a PD biomarker for biosimilarity assessment, such as a true dose–response relationship, relevance to mechanism of action, return to baseline post-treatment, sensitivity, and low variability. This finally resulted in the selection of three candidates (Lymphocyte activation gene 3 protein [LAG3], granulins [GRN], and CXC motif chemokine 11 [I-TAC]) that may have the greatest potential as PD biomarker for INFβ-1a biosimilars. It should be emphasized that not only is the reproducibility of the results of great importance, but upstream pathway analyses should also be performed to demonstrate the causal relationship between the respective biological drug and the identified biomarkers. This was the case for all three candidates mentioned above.

Uncertainty in regulatory acceptance has been a main barrier in the utilization of PD biomarkers for biosimilar development.5 The study by Chekka et al. is the first study that provides proof-of-concept that proteomics can be used to identify and develop PD biomarkers to demonstrate biosimilarity and provides a framework for how this can be applied across biosimilar development and approval.

No funding was received for this work.

The author declared no competing interests for this work.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
12.70
自引率
7.50%
发文量
290
审稿时长
2 months
期刊介绍: Clinical Pharmacology & Therapeutics (CPT) is the authoritative cross-disciplinary journal in experimental and clinical medicine devoted to publishing advances in the nature, action, efficacy, and evaluation of therapeutics. CPT welcomes original Articles in the emerging areas of translational, predictive and personalized medicine; new therapeutic modalities including gene and cell therapies; pharmacogenomics, proteomics and metabolomics; bioinformation and applied systems biology complementing areas of pharmacokinetics and pharmacodynamics, human investigation and clinical trials, pharmacovigilence, pharmacoepidemiology, pharmacometrics, and population pharmacology.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信