Yuli Qian, Louisa Schlachter, Doerthe Eckert, Sven Stodtmann, Anna Shmagel, Yi Peng, Wei Liu, Mohamed-Eslam F Mohamed
Juvenile idiopathic arthritis (JIA) is the most prevalent pediatric rheumatic disease. While disease-modifying antirheumatic drugs (DMARDs), especially biologics, have greatly transformed the management of JIA, there remain some unmet medical needs that require new treatment options. The objective of this work was to describe and apply a modeling and simulation approach to extrapolate upadacitinib efficacy from the adult diseases, rheumatoid arthritis (RA) and psoriatic arthritis (PsA), to their respective pediatric diseases, polyarticular course JIA (pcJIA), and juvenile PsA (JPsA). A population pharmacokinetic model characterized upadacitinib pharmacokinetics in pediatric patients using data from two phase I studies in pediatric patients with pcJIA (N = 51) or atopic dermatitis (N = 33). Efficacy simulations were conducted using previously developed exposure-response models in adults with RA and PsA. Real-world pcJIA and JPsA patient databases were leveraged to construct representative patient profiles for the targeted population. Following administration of the proposed weight-based dosing regimen, the model-predicted median upadacitinib plasma exposures in pediatric patients were within 20% of those in adult RA and PsA patients receiving the approved adult regimen. Simulations demonstrate that upadacitinib efficacy in pcJIA and JPsA is predicted to be non-inferior to that in adults with RA or PsA, respectively. The results of this work enabled recent approvals of upadacitinib for the treatment of polyarticular JIA and JPsA in the United States. Upadacitinib safety in pediatrics is being further evaluated in ongoing clinical trials.
幼年特发性关节炎(JIA)是发病率最高的儿科风湿病。虽然改变病情抗风湿药(DMARDs),尤其是生物制剂,已经大大改变了JIA的治疗方法,但仍有一些医疗需求未得到满足,需要新的治疗方案。这项工作的目的是描述并应用一种建模和模拟方法,将达帕替尼的疗效从成人疾病类风湿性关节炎(RA)和银屑病关节炎(PsA)推断到各自的儿科疾病多关节病程JIA(pcJIA)和幼年PsA(JPsA)。一个群体药代动力学模型利用在患有pcJIA(51例)或特应性皮炎(33例)的儿科患者中进行的两项I期研究的数据,描述了达达替尼在儿科患者中的药代动力学特征。疗效模拟是利用之前开发的成人 RA 和 PsA 暴露-反应模型进行的。利用真实世界的 pcJIA 和 JPsA 患者数据库,为目标人群建立了具有代表性的患者档案。在采用建议的基于体重的给药方案后,模型预测的儿科患者的中位upadacitinib血浆暴露量与接受已获批准的成人方案治疗的成人RA和PsA患者的暴露量相差不到20%。模拟结果表明,预计奥达帕替尼对pcJIA和JPsA的疗效分别不劣于对成人RA或PsA的疗效。这项工作的结果使美国最近批准了奥达替尼用于治疗多关节JIA和JPsA。目前正在进行的临床试验中进一步评估奥达替尼对儿科患者的安全性。
{"title":"Extrapolation of Upadacitinib Efficacy in Juvenile Idiopathic Arthritis Leveraging Pharmacokinetics, Exposure-Response Models, and Real-World Patient Data.","authors":"Yuli Qian, Louisa Schlachter, Doerthe Eckert, Sven Stodtmann, Anna Shmagel, Yi Peng, Wei Liu, Mohamed-Eslam F Mohamed","doi":"10.1002/cpt.3441","DOIUrl":"https://doi.org/10.1002/cpt.3441","url":null,"abstract":"<p><p>Juvenile idiopathic arthritis (JIA) is the most prevalent pediatric rheumatic disease. While disease-modifying antirheumatic drugs (DMARDs), especially biologics, have greatly transformed the management of JIA, there remain some unmet medical needs that require new treatment options. The objective of this work was to describe and apply a modeling and simulation approach to extrapolate upadacitinib efficacy from the adult diseases, rheumatoid arthritis (RA) and psoriatic arthritis (PsA), to their respective pediatric diseases, polyarticular course JIA (pcJIA), and juvenile PsA (JPsA). A population pharmacokinetic model characterized upadacitinib pharmacokinetics in pediatric patients using data from two phase I studies in pediatric patients with pcJIA (N = 51) or atopic dermatitis (N = 33). Efficacy simulations were conducted using previously developed exposure-response models in adults with RA and PsA. Real-world pcJIA and JPsA patient databases were leveraged to construct representative patient profiles for the targeted population. Following administration of the proposed weight-based dosing regimen, the model-predicted median upadacitinib plasma exposures in pediatric patients were within 20% of those in adult RA and PsA patients receiving the approved adult regimen. Simulations demonstrate that upadacitinib efficacy in pcJIA and JPsA is predicted to be non-inferior to that in adults with RA or PsA, respectively. The results of this work enabled recent approvals of upadacitinib for the treatment of polyarticular JIA and JPsA in the United States. Upadacitinib safety in pediatrics is being further evaluated in ongoing clinical trials.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.3,"publicationDate":"2024-09-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142337862","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Rare diseases, affecting millions globally, present significant drug development challenges. This is due to the limited patient populations and the unique pathophysiology of these diseases, which can make traditional clinical trial designs unfeasible. Quantitative Systems Pharmacology (QSP) models offer a promising approach to expedite drug development, particularly in rare diseases. QSP models provide a mechanistic representation of the disease and drug response in virtual patients that can complement routinely applied empirical modeling and simulation approaches. QSP models can generate digital twins of actual patients and mechanistically simulate the disease progression of rare diseases, accounting for phenotypic heterogeneity. QSP models can also support drug development in various drug modalities, such as gene therapy. Impactful QSP models case studies are presented here to illustrate their value in supporting various aspects of drug development in rare indications. As these QSP model applications continue to mature, there is a growing possibility that they could be more widely integrated into routine drug development steps. This integration could provide a robust framework for addressing some of the inherent challenges in rare disease drug development.
{"title":"Quantitative Systems Pharmacology Models: Potential Tools for Advancing Drug Development for Rare Diseases.","authors":"Susana Neves-Zaph, Chanchala Kaddi","doi":"10.1002/cpt.3451","DOIUrl":"https://doi.org/10.1002/cpt.3451","url":null,"abstract":"<p><p>Rare diseases, affecting millions globally, present significant drug development challenges. This is due to the limited patient populations and the unique pathophysiology of these diseases, which can make traditional clinical trial designs unfeasible. Quantitative Systems Pharmacology (QSP) models offer a promising approach to expedite drug development, particularly in rare diseases. QSP models provide a mechanistic representation of the disease and drug response in virtual patients that can complement routinely applied empirical modeling and simulation approaches. QSP models can generate digital twins of actual patients and mechanistically simulate the disease progression of rare diseases, accounting for phenotypic heterogeneity. QSP models can also support drug development in various drug modalities, such as gene therapy. Impactful QSP models case studies are presented here to illustrate their value in supporting various aspects of drug development in rare indications. As these QSP model applications continue to mature, there is a growing possibility that they could be more widely integrated into routine drug development steps. This integration could provide a robust framework for addressing some of the inherent challenges in rare disease drug development.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.3,"publicationDate":"2024-09-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142337867","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Chi-Chung Li, Brendan Bender, Justin Wilkins, Feifei Li, David C Turner, Bei Wang, Rong Deng, Shweta Vadhavkar, Zao Li, Antonia Kwan, Huang Huang, Kun Peng, Elicia Penuel, Ling-Yuh Huw, Pascal Chanu, Chunze Li, Shen Yin, Michael C Wei
Mosunetuzumab, a T-cell engaging bispecific antibody targeting CD20xCD3, is approved for treating relapsed/refractory follicular lymphoma. This research supports the approved intravenous clinical dose regimen, summarizing the exposure-response relationships for clinical safety and efficacy. A population pharmacokinetic model and Emax logistic regression exposure-response models for safety and efficacy were developed using data from 439 patients with relapsed/refractory non-Hodgkin lymphoma and 159 patients with relapsed/refractory follicular lymphoma, respectively, from a Phase I/II study (NCT02500407). Data from 0.2 to 60 mg across fixed dosing (Cohort A) and Cycle 1 step-up dosing (Cohort B) were used. Exposure-response models, using two-cycle area-under-the-concentration curve (AUC0-42) as the primary exposure endpoint, accurately depicted the complete response and objective response rate data across a 600-fold AUC0-42 range. The approved clinical dose regimen of 1/2/60/30 mg achieved near-maximal efficacy, with model-estimated CR and ORR (90% confidence interval) of 63.1% (49.7-75.0) and 79.1% (69.1-87.7), respectively. The exposure-response analysis for Grade ≥ 2 cytokine release syndrome identified receptor occupancy (%) within the first two cycles as a driver, with CRS dissipating beyond the first dosing cycle. No exposure-dependent increases were observed for other serious adverse events, including neutropenia and infections. The approved intravenous step-up dose regimen (i.e., step doses of 1 and 2 mg on Day 1 and 8, respectively) mitigated severe CRS risk, allowing safe administration of loading (60 mg) and target doses (30 mg every 3 weeks) to achieve a favorable benefit-risk profile.
{"title":"A Novel Step-Up Dosage Regimen for Enhancing the Benefit-to-Risk Ratio of Mosunetuzumab in Relapsed or Refractory Follicular Lymphoma.","authors":"Chi-Chung Li, Brendan Bender, Justin Wilkins, Feifei Li, David C Turner, Bei Wang, Rong Deng, Shweta Vadhavkar, Zao Li, Antonia Kwan, Huang Huang, Kun Peng, Elicia Penuel, Ling-Yuh Huw, Pascal Chanu, Chunze Li, Shen Yin, Michael C Wei","doi":"10.1002/cpt.3445","DOIUrl":"https://doi.org/10.1002/cpt.3445","url":null,"abstract":"<p><p>Mosunetuzumab, a T-cell engaging bispecific antibody targeting CD20xCD3, is approved for treating relapsed/refractory follicular lymphoma. This research supports the approved intravenous clinical dose regimen, summarizing the exposure-response relationships for clinical safety and efficacy. A population pharmacokinetic model and E<sub>max</sub> logistic regression exposure-response models for safety and efficacy were developed using data from 439 patients with relapsed/refractory non-Hodgkin lymphoma and 159 patients with relapsed/refractory follicular lymphoma, respectively, from a Phase I/II study (NCT02500407). Data from 0.2 to 60 mg across fixed dosing (Cohort A) and Cycle 1 step-up dosing (Cohort B) were used. Exposure-response models, using two-cycle area-under-the-concentration curve (AUC<sub>0-42</sub>) as the primary exposure endpoint, accurately depicted the complete response and objective response rate data across a 600-fold AUC<sub>0-42</sub> range. The approved clinical dose regimen of 1/2/60/30 mg achieved near-maximal efficacy, with model-estimated CR and ORR (90% confidence interval) of 63.1% (49.7-75.0) and 79.1% (69.1-87.7), respectively. The exposure-response analysis for Grade ≥ 2 cytokine release syndrome identified receptor occupancy (%) within the first two cycles as a driver, with CRS dissipating beyond the first dosing cycle. No exposure-dependent increases were observed for other serious adverse events, including neutropenia and infections. The approved intravenous step-up dose regimen (i.e., step doses of 1 and 2 mg on Day 1 and 8, respectively) mitigated severe CRS risk, allowing safe administration of loading (60 mg) and target doses (30 mg every 3 weeks) to achieve a favorable benefit-risk profile.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.3,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142337861","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Mufaddal Mahesri, Su Been Lee, Raisa Levin, Suzan Imren, Lanju Zhang, Timothy Beukelman, Lina Titievsky, Rishi J Desai
The CRISPR-based gene editing therapy exagamglogene autotemcel (exa-cel) recently received FDA approval for patients with severe sickle cell disease (SCD). The approval was based on a phase III trial (CLIMB SCD 121), which showed 97% efficacy of this treatment in eliminating vaso occlusive crises (VOCs) for 12 consecutive months. To help contextualize results from this trial, we aimed to investigate the proportion of patients with severe SCD who remain VOC-free for a 1-year period in routine clinical care. Using Medicaid claims data (2000-2018), we identified a cohort of patients, 12-35 years old with severe SCD, defined by ≥ 2 VOCs per year for 2 consecutive years, who met other exa-cel trial inclusion criteria to mimic a trial-like population. A VOC was identified using ICD diagnosis codes during hospitalization and ER visits. The primary outcome was the proportion of patients with no VOCs during a 1-year follow-up. A total of 7,425 patients with severe SCD [mean (SD) age: 20.5 (6.0) years, 54.6% females, 84% African Americans], had a mean of 5.2 VOCs, 5.1 ER visits and 3.5 hospitalizations per year during the baseline period. The proportion of patients with no VOCs during the 1-year follow-up was 7.7% (95% confidence interval: 7.1%-8.3%). In conclusion, less than one in 12 patients with severe SCD achieved VOC-free status within 1 year in routine clinical care. These findings suggest that the high efficacy observed for exa-cel in the trial, if replicated in routine clinical care, could translate into a significant public health impact.
{"title":"Infrequent Resolution of Vaso-Occlusive Crises in Routine Clinical Care Among Patients Mimicking the Exa-Cel Trial Population: A Cohort Study of Medicaid Enrollees.","authors":"Mufaddal Mahesri, Su Been Lee, Raisa Levin, Suzan Imren, Lanju Zhang, Timothy Beukelman, Lina Titievsky, Rishi J Desai","doi":"10.1002/cpt.3449","DOIUrl":"https://doi.org/10.1002/cpt.3449","url":null,"abstract":"<p><p>The CRISPR-based gene editing therapy exagamglogene autotemcel (exa-cel) recently received FDA approval for patients with severe sickle cell disease (SCD). The approval was based on a phase III trial (CLIMB SCD 121), which showed 97% efficacy of this treatment in eliminating vaso occlusive crises (VOCs) for 12 consecutive months. To help contextualize results from this trial, we aimed to investigate the proportion of patients with severe SCD who remain VOC-free for a 1-year period in routine clinical care. Using Medicaid claims data (2000-2018), we identified a cohort of patients, 12-35 years old with severe SCD, defined by ≥ 2 VOCs per year for 2 consecutive years, who met other exa-cel trial inclusion criteria to mimic a trial-like population. A VOC was identified using ICD diagnosis codes during hospitalization and ER visits. The primary outcome was the proportion of patients with no VOCs during a 1-year follow-up. A total of 7,425 patients with severe SCD [mean (SD) age: 20.5 (6.0) years, 54.6% females, 84% African Americans], had a mean of 5.2 VOCs, 5.1 ER visits and 3.5 hospitalizations per year during the baseline period. The proportion of patients with no VOCs during the 1-year follow-up was 7.7% (95% confidence interval: 7.1%-8.3%). In conclusion, less than one in 12 patients with severe SCD achieved VOC-free status within 1 year in routine clinical care. These findings suggest that the high efficacy observed for exa-cel in the trial, if replicated in routine clinical care, could translate into a significant public health impact.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.3,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142337864","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"The Promise of Health for All: A Collaborative Model to Close Treatment Gaps for Neglected Diseases.","authors":"Delali Attipoe","doi":"10.1002/cpt.3427","DOIUrl":"https://doi.org/10.1002/cpt.3427","url":null,"abstract":"","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.3,"publicationDate":"2024-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142337869","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Noah C Neverette, Julie B Dumond, Deborah K McMahon, Aaron S Devanathan
The mainstay of antiretroviral therapy (ART) has been combination oral therapy. While oral ART is highly effective, nonadherence remains a chief concern. Addressing this concern in recent years is the emergence of long-acting antiretrovirals for the treatment and prevention of HIV-1 infection. The most recently approved long-acting antiretroviral is the first-in-class capsid inhibitor lenacapavir (LEN) for heavily treatment-experienced adults with multidrug-resistant HIV-1 infection. Due to its biannual subcutaneous dosing scheme to inhibit the HIV-1 capsid, LEN exhibits unique pharmacokinetics and reinforces an evolving era of ART. In this review, we evaluate published and accepted research articles, conference proceedings, and clinical trial records to provide a comprehensive overview of LEN for treatment and preliminary data for the prevention of HIV-1 infection. These data include clinical trials outcomes, in vitro and in vivo resistance profiles, and preclinical data supporting downstream indications. We also discuss the unique clinical pharmacology of LEN with the goal of serving as a resource toward subsequent physiologically based, population-based, and other miscellaneous pharmacometric-focused analyses. Given the dynamic nature of the HIV treatment and prevention research fields, we also discuss ongoing studies related to LEN for treatment-naïve adults and for prevention. Lastly, we discuss important pharmacologic gaps in special populations, drug-drug interactions, and at the sites of action germane to HIV treatment and prevention. The information discussed in this review will provide knowledge and understanding of the unique pharmacologic properties of LEN to assist clinicians and researchers as they navigate the dynamic HIV research landscape.
抗逆转录病毒疗法(ART)的主流是口服联合疗法。虽然口服抗逆转录病毒疗法非常有效,但不坚持治疗仍是一个主要问题。为了解决这一问题,近年来出现了用于治疗和预防 HIV-1 感染的长效抗逆转录病毒药物。最近获批的长效抗逆转录病毒药物是第一类胶囊抑制剂来那卡韦(LEN),用于治疗耐多药HIV-1感染的有大量治疗经验的成人患者。由于来那卡韦采用一年两次的皮下给药方案来抑制HIV-1衣壳,因此它表现出独特的药代动力学,巩固了不断发展的抗逆转录病毒疗法时代。在这篇综述中,我们评估了已发表和接受的研究文章、会议记录和临床试验记录,全面概述了用于治疗的 LEN 以及用于预防 HIV-1 感染的初步数据。这些数据包括临床试验结果、体外和体内耐药性特征以及支持下游适应症的临床前数据。我们还讨论了 LEN 独特的临床药理学,目的是为后续的生理学分析、基于人群的分析以及其他以药效学为重点的分析提供资源。鉴于 HIV 治疗和预防研究领域的动态性质,我们还讨论了正在进行的与 LEN 有关的研究,这些研究针对治疗无效的成人和预防。最后,我们讨论了在特殊人群、药物间相互作用以及与 HIV 治疗和预防相关的作用位点方面存在的重要药理差距。本综述中讨论的信息将为临床医生和研究人员提供有关 LEN 独特药理特性的知识和理解,帮助他们驾驭充满活力的 HIV 研究环境。
{"title":"Lenacapavir: Playing the Long Game in the New Era of Antiretrovirals.","authors":"Noah C Neverette, Julie B Dumond, Deborah K McMahon, Aaron S Devanathan","doi":"10.1002/cpt.3447","DOIUrl":"https://doi.org/10.1002/cpt.3447","url":null,"abstract":"<p><p>The mainstay of antiretroviral therapy (ART) has been combination oral therapy. While oral ART is highly effective, nonadherence remains a chief concern. Addressing this concern in recent years is the emergence of long-acting antiretrovirals for the treatment and prevention of HIV-1 infection. The most recently approved long-acting antiretroviral is the first-in-class capsid inhibitor lenacapavir (LEN) for heavily treatment-experienced adults with multidrug-resistant HIV-1 infection. Due to its biannual subcutaneous dosing scheme to inhibit the HIV-1 capsid, LEN exhibits unique pharmacokinetics and reinforces an evolving era of ART. In this review, we evaluate published and accepted research articles, conference proceedings, and clinical trial records to provide a comprehensive overview of LEN for treatment and preliminary data for the prevention of HIV-1 infection. These data include clinical trials outcomes, in vitro and in vivo resistance profiles, and preclinical data supporting downstream indications. We also discuss the unique clinical pharmacology of LEN with the goal of serving as a resource toward subsequent physiologically based, population-based, and other miscellaneous pharmacometric-focused analyses. Given the dynamic nature of the HIV treatment and prevention research fields, we also discuss ongoing studies related to LEN for treatment-naïve adults and for prevention. Lastly, we discuss important pharmacologic gaps in special populations, drug-drug interactions, and at the sites of action germane to HIV treatment and prevention. The information discussed in this review will provide knowledge and understanding of the unique pharmacologic properties of LEN to assist clinicians and researchers as they navigate the dynamic HIV research landscape.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.3,"publicationDate":"2024-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142337865","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jaime Algorta, Stella Kepha, Alejandro Krolewiecki, Hanbin Li, Justin Giang, Pedro Fleitas, Charles Mwandawiro, José Muñoz
Trichuris trichiura is a soil-transmitted helminth causing intestinal disease. Albendazole is the standard treatment despite its moderate efficacy, which is improved when co-administered with ivermectin. A fixed-dose combination adds practical advantages mainly for mass drug administration. The aim of this article is to define the population pharmacokinetic models and exposure-response of an innovative albendazole/ivermectin combination. Data were obtained from a phase I clinical trial in healthy adults and from a phase II trial in children and adolescents infected with T. trichiura. Nonlinear mixed-effects models were built for albendazole and ivermectin using NONMEM®. Area under the curve was calculated using the empirical Bayes estimates of the pharmacokinetic parameters of each individual and used for evaluation of exposure-response between cure rate and pharmacokinetic exposure. The pharmacokinetics of albendazole was described using a two-compartmental model with first-order absorption and the pharmacokinetics of ivermectin was described using a two-compartmental model with zero-order followed by first-order absorption. Clearance and volume of distribution increased with body weight for both albendazole and ivermectin. Day 1 area under the curve of albendazole and ivermectin from the children and adolescents treated with the combination regimens were similar to the healthy adults treated with control drugs. A flat exposure-response relationship was observed between the cure rate and drug exposure. Population pharmacokinetic of a combination of albendazole and ivermectin in children, adolescents, and adults, either healthy or infected by T. trichiura was described. The dosage selected in the phase II trial was appropriate for the subsequent phase III.
毛滴虫是一种通过土壤传播的蠕虫,可引起肠道疾病。阿苯达唑是标准的治疗方法,尽管其疗效一般,但与伊维菌素合用可提高疗效。固定剂量复方制剂主要在大规模用药方面更具实用优势。本文旨在确定创新型阿苯达唑/伊维菌素复方制剂的群体药代动力学模型和暴露-反应。数据来源于一项针对健康成人的 I 期临床试验和一项针对感染滴虫的儿童和青少年的 II 期试验。使用 NONMEM® 为阿苯达唑和伊维菌素建立了非线性混合效应模型。使用每个个体的药代动力学参数的经验贝叶斯估计值计算曲线下面积,并用于评估治愈率和药代动力学暴露量之间的暴露-反应。阿苯达唑的药代动力学采用一阶吸收的二室模型进行描述,伊维菌素的药代动力学采用先零阶后一阶吸收的二室模型进行描述。阿苯达唑和伊维菌素的清除率和分布容积均随体重增加而增加。使用联合疗法治疗的儿童和青少年体内阿苯达唑和伊维菌素的第一天曲线下面积与使用对照药物治疗的健康成人相似。在治愈率和药物暴露量之间观察到一种平坦的暴露量-反应关系。该研究描述了阿苯达唑和伊维菌素复方制剂在儿童、青少年和成人(健康或感染了毛滴虫)中的群体药代动力学。在 II 期试验中选择的剂量适合随后的 III 期试验。
{"title":"Population Pharmacokinetics and Exposure-Response Analysis of a Fixed-Dose Combination of Ivermectin and Albendazole in Children, Adolescents, and Adults.","authors":"Jaime Algorta, Stella Kepha, Alejandro Krolewiecki, Hanbin Li, Justin Giang, Pedro Fleitas, Charles Mwandawiro, José Muñoz","doi":"10.1002/cpt.3424","DOIUrl":"https://doi.org/10.1002/cpt.3424","url":null,"abstract":"<p><p>Trichuris trichiura is a soil-transmitted helminth causing intestinal disease. Albendazole is the standard treatment despite its moderate efficacy, which is improved when co-administered with ivermectin. A fixed-dose combination adds practical advantages mainly for mass drug administration. The aim of this article is to define the population pharmacokinetic models and exposure-response of an innovative albendazole/ivermectin combination. Data were obtained from a phase I clinical trial in healthy adults and from a phase II trial in children and adolescents infected with T. trichiura. Nonlinear mixed-effects models were built for albendazole and ivermectin using NONMEM®. Area under the curve was calculated using the empirical Bayes estimates of the pharmacokinetic parameters of each individual and used for evaluation of exposure-response between cure rate and pharmacokinetic exposure. The pharmacokinetics of albendazole was described using a two-compartmental model with first-order absorption and the pharmacokinetics of ivermectin was described using a two-compartmental model with zero-order followed by first-order absorption. Clearance and volume of distribution increased with body weight for both albendazole and ivermectin. Day 1 area under the curve of albendazole and ivermectin from the children and adolescents treated with the combination regimens were similar to the healthy adults treated with control drugs. A flat exposure-response relationship was observed between the cure rate and drug exposure. Population pharmacokinetic of a combination of albendazole and ivermectin in children, adolescents, and adults, either healthy or infected by T. trichiura was described. The dosage selected in the phase II trial was appropriate for the subsequent phase III.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.3,"publicationDate":"2024-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142277588","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Gauri G Rao, Quentin Vallé, Ramya Mahadevan, Rajnikant Sharma, Jeremy J Barr, Daria Van Tyne
Effectively treating multidrug-resistant bacterial infections remains challenging due to the limited drug development pipeline and a scarcity of novel agents effective against these highly resistant pathogens. Bacteriophages (phages) are a potential addition to the antimicrobial treatment arsenal. Though, phages are currently being tested in clinical trials for antibiotic-resistant infections, phages lack a fundamental understanding of optimal dosing in humans. Rationally designed preclinical studies using in vitro and in vivo infection models, allow us to assess clinically relevant phage +/- antibiotic exposure (pharmacokinetics), the resulting treatment impact on the infecting pathogen (pharmacodynamics) and host immune response (immunodynamics). A mechanistic modeling framework allows us to integrate this knowledge gained from preclinical studies to develop predictive models. We reviewed recently published mathematical models based on in vitro and/or in vivo data that evaluate the effects of varying bacterial or phage densities, phage characteristics (burst size, adsorption rate), phage pharmacokinetics, phage-antibiotic combinations and host immune responses. In our review, we analyzed study designs and the data used to inform the development of these mechanistic models. Insights gained from model-based simulations were reviewed as they help identify crucial phage parameters for determining effective phage dosing. These efforts contribute to bridging the gap between phage therapy research and its clinical translation.
{"title":"Crossing the Chasm: How to Approach Translational Pharmacokinetic-Pharmacodynamic Modeling of Phage Dosing.","authors":"Gauri G Rao, Quentin Vallé, Ramya Mahadevan, Rajnikant Sharma, Jeremy J Barr, Daria Van Tyne","doi":"10.1002/cpt.3426","DOIUrl":"https://doi.org/10.1002/cpt.3426","url":null,"abstract":"<p><p>Effectively treating multidrug-resistant bacterial infections remains challenging due to the limited drug development pipeline and a scarcity of novel agents effective against these highly resistant pathogens. Bacteriophages (phages) are a potential addition to the antimicrobial treatment arsenal. Though, phages are currently being tested in clinical trials for antibiotic-resistant infections, phages lack a fundamental understanding of optimal dosing in humans. Rationally designed preclinical studies using in vitro and in vivo infection models, allow us to assess clinically relevant phage +/- antibiotic exposure (pharmacokinetics), the resulting treatment impact on the infecting pathogen (pharmacodynamics) and host immune response (immunodynamics). A mechanistic modeling framework allows us to integrate this knowledge gained from preclinical studies to develop predictive models. We reviewed recently published mathematical models based on in vitro and/or in vivo data that evaluate the effects of varying bacterial or phage densities, phage characteristics (burst size, adsorption rate), phage pharmacokinetics, phage-antibiotic combinations and host immune responses. In our review, we analyzed study designs and the data used to inform the development of these mechanistic models. Insights gained from model-based simulations were reviewed as they help identify crucial phage parameters for determining effective phage dosing. These efforts contribute to bridging the gap between phage therapy research and its clinical translation.</p>","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.3,"publicationDate":"2024-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142306776","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
T-cell-engaging bispecific antibodies (TCEs) that target tumor antigens and T cells have shown great promise in treating cancer, particularly in hematological indications. The clinical development of TCEs often involves a lengthy first-in-human (FIH) trial with many dose-escalation cohorts leading up to an early proof of concept (POC), enabling either a no-go decision or dose selection for further clinical development. Multiple factors related to the target, product, disease, and patient population influence the efficacy and safety of TCEs. The intricate mechanism of action limits the translatability of preclinical models to the clinic, thereby posing challenges to streamline clinical development. In addition, unlike traditional chemotherapy, the top dose and recommended phase II doses (RP2Ds) for TCEs in the clinic are often not guided by the maximum tolerated dose (MTD), but rather based on the integrated dose-response assessment of the benefit/risk profile. These uncertainties pose complex challenges for translational and clinical pharmacologists (PK/PD scientists), as well as clinicians, to design an efficient clinical study that guides development. To that end, experts in the field, under the umbrella of the American Association of Pharmaceutical Scientists, have reviewed learnings from published literature and currently marketed products to share perspectives on the FIH and clinical pharmacology strategies to support early clinical development of TCEs.
靶向肿瘤抗原和 T 细胞的 T 细胞靶向双特异性抗体 (TCE) 在治疗癌症,尤其是血液病适应症方面大有可为。双特异性抗体的临床开发通常需要经过漫长的首次人体试验(FIH)和多次剂量递增试验,最终获得早期概念验证(POC),从而决定是否进行进一步的临床开发或选择剂量。与靶点、产品、疾病和患者群体有关的多种因素会影响 TCE 的疗效和安全性。复杂的作用机制限制了临床前模型向临床的转化,从而为简化临床开发带来了挑战。此外,与传统化疗不同,TCEs 在临床中的最高剂量和二期推荐剂量(RP2D)往往不是以最大耐受剂量(MTD)为指导,而是基于对获益/风险概况的综合剂量-反应评估。这些不确定性给转化和临床药理学家(PK/PD 科学家)以及临床医生带来了复杂的挑战,如何才能设计出高效的临床研究来指导研发工作?为此,该领域的专家们在美国医药科学家协会的支持下,回顾了从已发表的文献和目前上市的产品中学到的知识,分享了对 FIH 和临床药理策略的看法,以支持 TCE 的早期临床开发。
{"title":"Industry Perspective on First-in-Human and Clinical Pharmacology Strategies to Support Clinical Development of T-Cell Engaging Bispecific Antibodies for Cancer Therapy.","authors":"Prathap Nagaraja Shastri,Nirav Shah,Martin Lechmann,Hardik Mody,Marc W Retter,Min Zhu,Tommy Li,Jun Wang,Naveed Shaik,Xirong Zheng,Meric Ovacik,Fei Hua,Vibha Jawa,Christophe Boetsch,Yanguang Cao,John Burke,Kaushik Datta,Kapil Gadkar,Vijay Upreti,Alison Betts","doi":"10.1002/cpt.3439","DOIUrl":"https://doi.org/10.1002/cpt.3439","url":null,"abstract":"T-cell-engaging bispecific antibodies (TCEs) that target tumor antigens and T cells have shown great promise in treating cancer, particularly in hematological indications. The clinical development of TCEs often involves a lengthy first-in-human (FIH) trial with many dose-escalation cohorts leading up to an early proof of concept (POC), enabling either a no-go decision or dose selection for further clinical development. Multiple factors related to the target, product, disease, and patient population influence the efficacy and safety of TCEs. The intricate mechanism of action limits the translatability of preclinical models to the clinic, thereby posing challenges to streamline clinical development. In addition, unlike traditional chemotherapy, the top dose and recommended phase II doses (RP2Ds) for TCEs in the clinic are often not guided by the maximum tolerated dose (MTD), but rather based on the integrated dose-response assessment of the benefit/risk profile. These uncertainties pose complex challenges for translational and clinical pharmacologists (PK/PD scientists), as well as clinicians, to design an efficient clinical study that guides development. To that end, experts in the field, under the umbrella of the American Association of Pharmaceutical Scientists, have reviewed learnings from published literature and currently marketed products to share perspectives on the FIH and clinical pharmacology strategies to support early clinical development of TCEs.","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.7,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142260299","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"What Is Rare and What Is Orphan? A Guide to the Regulatory Terminology","authors":"Kristina Larsson","doi":"10.1002/cpt.3446","DOIUrl":"https://doi.org/10.1002/cpt.3446","url":null,"abstract":"","PeriodicalId":153,"journal":{"name":"Clinical Pharmacology & Therapeutics","volume":null,"pages":null},"PeriodicalIF":6.7,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142269874","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}