First-in-Human Dose Selection for T-Cell Engaging Bispecific Antibodies

IF 6.3 2区 医学 Q1 PHARMACOLOGY & PHARMACY
Piet H. van der Graaf
{"title":"First-in-Human Dose Selection for T-Cell Engaging Bispecific Antibodies","authors":"Piet H. van der Graaf","doi":"10.1002/cpt.3487","DOIUrl":null,"url":null,"abstract":"<p>Although bispecific antibodies (“bispecifics,” antibodies that can bind to two different antigens at the same time) are typically coined as novel modalities,<span><sup>1</sup></span> they were already reported in 1960 by Nisonoff and colleagues.<span><sup>2</sup></span> However, it was only three decades later for the first clinical investigation to happen and then it took another two to the approval of the first bispecific antibody for human therapeutic use.<span><sup>3</sup></span> There are now more than 10 bispecific antibodies approved by the US Food and Drug Administration (FDA), the majority for the treatment of cancer (<b>Figure</b> 1).<span><sup>4</sup></span></p><p>The initial steps and milestones in the development of bispecific therapeutics were mainly fueled by advances and breakthroughs in biotechnology and antibody engineering. More recently biology, translational medicine as well as clinical pharmacology have played an increasingly important role. The most exciting properties of bispecifics are called obligate, since they are dependent on the physical linkage of the two “arms” and cannot be obtained by combining separate antibodies with the same individual binding properties.<span><sup>3</sup></span> An example is avidity (sometimes referred to as functional affinity), which quantifies the synergy due to multiple simultaneous binding interactions.<span><sup>3, 5</sup></span> These unique properties of bispecifics offer potential therapeutic advantages but also raise significant drug development challenges. For example, the dose/concentration-response relationship for bispecifics can be bell-shaped, which makes dose-finding more challenging.<span><sup>5</sup></span> Second, due to highly system-dependent pharmacology, the translation of pharmacokinetics-pharmacodynamics (PKPD) of bispecifics is complex and traditional minimal anticipated biological effect level (MABEL) concepts to define first-in-human (FIH) starting dose for conventional monoclonal antibodies do not apply.<span><sup>6</sup></span> Last but not least, how do we predict and manage between-patient variability, which due to the multifactorial mechanism of action may be more pronounced for bispecifics than what is typically observed with conventional monoclonal antibodies.<span><sup>5</sup></span></p><p>A series of recent white papers, reviews, and research articles published in <i>Clinical Pharmacology &amp; Therapeutics</i> (<i>CPT</i>) reflects the significant interest of drug developers in this area and provides overviews of current clinical pharmacology best practices and innovation.<span><sup>4, 7-11</sup></span> In this issue of <i>CPT</i>, a working group from the <i>American Association of Pharmaceutical Scientists</i> (<i>AAPS</i>) presents learnings and recommendations for FIH starting dose selection, early clinical development, and recommended Phase 2 dose (RP2D) selection and bioanalytical and biomarkers strategies for T-cell engaging bispecific antibodies.<span><sup>12</sup></span> What sets this White Paper apart from somewhat similar recent reviews is that it provides detailed recommendations on <i>in vitro</i> pharmacological assays to determine clinically relevant MABEL, and the authors “recommend defining MABEL at EC<sub>50</sub> using pharmacologically relevant MABEL assay setup.”<span><sup>12</sup></span> In a second paper, in this issue of <i>CPT</i>, Zhou, and coworkers try to address what a “pharmacologically relevant MABEL assay setup” is.<span><sup>13</sup></span> To guide FIH dose selection for the bispecific AMG 199, they lowered the effector to target (E:T) ratio in the most sensitive MABEL assay to better reflect the physiological circumstances of the gastric tumor microenvironment in patients. As a result, a 10-fold higher starting dose was selected compared to the one derived from the original assay, and the former was accepted by regulatory agencies. The higher starting dose saved at least two cohorts in FIH and the first clinical observations confirmed it was safe and well tolerated. The novel approach has now been validated across similar programs and the authors propose it can be applied to other bispecifics for both solid and hematological tumors.<span><sup>13</sup></span> An alternative strategy to optimize FIH starting dose for multi-specific biologics and avoid exposing cancer patients to sub-efficacious is the use of quantitative systems pharmacology (QSP) modeling, as discussed in a recent Editorial in this journal.<span><sup>14</sup></span> For example, Carretero-Iglesia <i>et al</i>. recently reported that a QSP approach resulted in a 50–100-fold higher starting dose for the trispecific ISB 2001 in multiple myeloma patients.<span><sup>15</sup></span> Similar examples<span><sup>12</sup></span> demonstrate that innovation in clinical pharmacology has a direct and tangible impact on the speed and cost of the development of bispecific therapeutics, and most importantly on patient lives.</p><p>No funding was received for this work.</p><p>As employee of Certara, PHvdG was involved in the support of the ISB 2001 program.<span><sup>15</sup></span></p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":"117 1","pages":"11-13"},"PeriodicalIF":6.3000,"publicationDate":"2024-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cpt.3487","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Clinical Pharmacology & Therapeutics","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cpt.3487","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

Although bispecific antibodies (“bispecifics,” antibodies that can bind to two different antigens at the same time) are typically coined as novel modalities,1 they were already reported in 1960 by Nisonoff and colleagues.2 However, it was only three decades later for the first clinical investigation to happen and then it took another two to the approval of the first bispecific antibody for human therapeutic use.3 There are now more than 10 bispecific antibodies approved by the US Food and Drug Administration (FDA), the majority for the treatment of cancer (Figure 1).4

The initial steps and milestones in the development of bispecific therapeutics were mainly fueled by advances and breakthroughs in biotechnology and antibody engineering. More recently biology, translational medicine as well as clinical pharmacology have played an increasingly important role. The most exciting properties of bispecifics are called obligate, since they are dependent on the physical linkage of the two “arms” and cannot be obtained by combining separate antibodies with the same individual binding properties.3 An example is avidity (sometimes referred to as functional affinity), which quantifies the synergy due to multiple simultaneous binding interactions.3, 5 These unique properties of bispecifics offer potential therapeutic advantages but also raise significant drug development challenges. For example, the dose/concentration-response relationship for bispecifics can be bell-shaped, which makes dose-finding more challenging.5 Second, due to highly system-dependent pharmacology, the translation of pharmacokinetics-pharmacodynamics (PKPD) of bispecifics is complex and traditional minimal anticipated biological effect level (MABEL) concepts to define first-in-human (FIH) starting dose for conventional monoclonal antibodies do not apply.6 Last but not least, how do we predict and manage between-patient variability, which due to the multifactorial mechanism of action may be more pronounced for bispecifics than what is typically observed with conventional monoclonal antibodies.5

A series of recent white papers, reviews, and research articles published in Clinical Pharmacology & Therapeutics (CPT) reflects the significant interest of drug developers in this area and provides overviews of current clinical pharmacology best practices and innovation.4, 7-11 In this issue of CPT, a working group from the American Association of Pharmaceutical Scientists (AAPS) presents learnings and recommendations for FIH starting dose selection, early clinical development, and recommended Phase 2 dose (RP2D) selection and bioanalytical and biomarkers strategies for T-cell engaging bispecific antibodies.12 What sets this White Paper apart from somewhat similar recent reviews is that it provides detailed recommendations on in vitro pharmacological assays to determine clinically relevant MABEL, and the authors “recommend defining MABEL at EC50 using pharmacologically relevant MABEL assay setup.”12 In a second paper, in this issue of CPT, Zhou, and coworkers try to address what a “pharmacologically relevant MABEL assay setup” is.13 To guide FIH dose selection for the bispecific AMG 199, they lowered the effector to target (E:T) ratio in the most sensitive MABEL assay to better reflect the physiological circumstances of the gastric tumor microenvironment in patients. As a result, a 10-fold higher starting dose was selected compared to the one derived from the original assay, and the former was accepted by regulatory agencies. The higher starting dose saved at least two cohorts in FIH and the first clinical observations confirmed it was safe and well tolerated. The novel approach has now been validated across similar programs and the authors propose it can be applied to other bispecifics for both solid and hematological tumors.13 An alternative strategy to optimize FIH starting dose for multi-specific biologics and avoid exposing cancer patients to sub-efficacious is the use of quantitative systems pharmacology (QSP) modeling, as discussed in a recent Editorial in this journal.14 For example, Carretero-Iglesia et al. recently reported that a QSP approach resulted in a 50–100-fold higher starting dose for the trispecific ISB 2001 in multiple myeloma patients.15 Similar examples12 demonstrate that innovation in clinical pharmacology has a direct and tangible impact on the speed and cost of the development of bispecific therapeutics, and most importantly on patient lives.

No funding was received for this work.

As employee of Certara, PHvdG was involved in the support of the ISB 2001 program.15

Abstract Image

t细胞结合双特异性抗体的首次人体剂量选择。
虽然双特异性抗体(“双特异性”,可以同时结合两种不同抗原的抗体)通常被认为是一种新形式,但早在1960年,Nisonoff和他的同事就已经报道了这种抗体然而,仅仅在30年后才进行了第一次临床研究,然后又花了2年时间才批准了第一个用于人类治疗的双特异性抗体目前,美国食品和药物管理局(FDA)批准了10多种双特异性抗体,其中大多数用于癌症治疗(图1)。双特异性治疗发展的最初步骤和里程碑主要是由生物技术和抗体工程的进步和突破推动的。最近,生物学、转化医学以及临床药理学发挥了越来越重要的作用。双特异性最令人兴奋的特性被称为专性,因为它们依赖于两个“臂”的物理连接,不能通过结合具有相同个体结合特性的单独抗体来获得一个例子是亲和度(有时称为功能亲和度),它量化了由于多个同时结合相互作用而产生的协同作用。3,5双特异性的这些独特性质提供了潜在的治疗优势,但也提出了重大的药物开发挑战。例如,双特异性的剂量/浓度-反应关系可能呈钟形,这使得剂量查找更具挑战性其次,由于高度系统依赖性的药理学,双特异性的药代动力学-药效学(PKPD)的翻译是复杂的,传统的最小预期生物效应水平(MABEL)概念来定义传统单克隆抗体的首次人体(FIH)起始剂量并不适用最后但并非最不重要的是,我们如何预测和管理患者之间的可变性,由于多因子作用机制,双特异性可能比传统单克隆抗体通常观察到的更为明显。《临床药理学》杂志近期发表的5A系列白皮书、综述和研究文章;治疗学(CPT)反映了药物开发人员在这一领域的重大兴趣,并提供了当前临床药理学最佳实践和创新的概述。在这一期的CPT中,来自美国制药科学家协会(AAPS)的一个工作组介绍了关于FIH起始剂量选择、早期临床开发、推荐的2期剂量(RP2D)选择以及t细胞双特异性抗体的生物分析和生物标志物策略的学习和建议本白皮书与最近类似的评论不同之处在于,它提供了关于体外药理学分析的详细建议,以确定临床相关的MABEL,作者“建议使用药理学相关的MABEL测定装置在EC50定义MABEL。”在这一期《CPT》的第二篇论文中,Zhou及其同事试图说明什么是“与药理学相关的MABEL测定装置”为了指导双特异性AMG 199的FIH剂量选择,他们在最敏感的MABEL试验中降低了效应靶比(E:T),以更好地反映患者胃肿瘤微环境的生理情况。结果,选择的起始剂量比从原始分析得出的剂量高10倍,并且前者被监管机构接受。较高的起始剂量在FIH中至少挽救了两个队列,并且首次临床观察证实它是安全且耐受性良好的。这种新方法现在已经在类似的项目中得到了验证,作者提出它可以应用于实体肿瘤和血液肿瘤的其他双特异性治疗正如本杂志最近的一篇社论所讨论的那样,优化多特异性生物制剂的FIH起始剂量并避免癌症患者暴露于无效的另一种策略是使用定量系统药理学(QSP)模型例如,Carretero-Iglesia等人最近报道,QSP方法可使三特异性ISB 2001在多发性骨髓瘤患者中的起始剂量提高50 - 100倍类似的例子表明,临床药理学的创新对双特异性治疗的发展速度和成本,最重要的是对病人的生命,有着直接而切实的影响。这项工作没有收到任何资金。作为Certara的员工,PHvdG参与了ISB 2001项目的支持
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
CiteScore
12.70
自引率
7.50%
发文量
290
审稿时长
2 months
期刊介绍: Clinical Pharmacology & Therapeutics (CPT) is the authoritative cross-disciplinary journal in experimental and clinical medicine devoted to publishing advances in the nature, action, efficacy, and evaluation of therapeutics. CPT welcomes original Articles in the emerging areas of translational, predictive and personalized medicine; new therapeutic modalities including gene and cell therapies; pharmacogenomics, proteomics and metabolomics; bioinformation and applied systems biology complementing areas of pharmacokinetics and pharmacodynamics, human investigation and clinical trials, pharmacovigilence, pharmacoepidemiology, pharmacometrics, and population pharmacology.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信