Courtney Moc Willeford, Krithika Shetty, Douglas Sheridan, Frank Engler
RLYB212 is a human monoclonal anti-human platelet antigen (HPA)-1a immunoglobulin gamma 1 in clinical development as a subcutaneous injection for the prevention of maternal alloimmunization to fetal HPA-1a leading to fetal and neonatal alloimmune thrombocytopenia (FNAIT). This analysis developed a target-mediated drug disposition (TMDD) model to simultaneously characterize RLYB212 pharmacokinetics (PK) and HPA-1a-positive platelet dynamics in HPA-1b/b (HPA-1a-negative) volunteers. The model was then used to perform simulations to inform a dosing regimen in a phase II clinical study in pregnant women, where simulations accounted for physiological changes throughout pregnancy. Allometric scaling (0.75) for clearance and intercompartment transfer rate and volume (1) was included in the base model to account for variations in body weight. A 0.06 mg RLYB212 dose with a loading dose of 0.12 mg was identified as the optimal dosing regimen of RLYB212, which maintained exposures below the target upper boundary of ~10 ng/mL throughout pregnancy. This work presents an application of the TMDD model that advances the quantitative clinical pharmacology toolkit to understand monoclonal antibody PK in pregnancy.
{"title":"Informing pregnancy dose via target-mediated drug disposition modeling and simulations for a recombinant human monoclonal antibody","authors":"Courtney Moc Willeford, Krithika Shetty, Douglas Sheridan, Frank Engler","doi":"10.1002/psp4.13250","DOIUrl":"10.1002/psp4.13250","url":null,"abstract":"<p>RLYB212 is a human monoclonal anti-human platelet antigen (HPA)-1a immunoglobulin gamma 1 in clinical development as a subcutaneous injection for the prevention of maternal alloimmunization to fetal HPA-1a leading to fetal and neonatal alloimmune thrombocytopenia (FNAIT). This analysis developed a target-mediated drug disposition (TMDD) model to simultaneously characterize RLYB212 pharmacokinetics (PK) and HPA-1a-positive platelet dynamics in HPA-1b/b (HPA-1a-negative) volunteers. The model was then used to perform simulations to inform a dosing regimen in a phase II clinical study in pregnant women, where simulations accounted for physiological changes throughout pregnancy. Allometric scaling (0.75) for clearance and intercompartment transfer rate and volume (1) was included in the base model to account for variations in body weight. A 0.06 mg RLYB212 dose with a loading dose of 0.12 mg was identified as the optimal dosing regimen of RLYB212, which maintained exposures below the target upper boundary of ~10 ng/mL throughout pregnancy. This work presents an application of the TMDD model that advances the quantitative clinical pharmacology toolkit to understand monoclonal antibody PK in pregnancy.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"13 11","pages":"2002-2015"},"PeriodicalIF":3.1,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.13250","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142575043","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Craig M Comisar, Jose Francis, Jim H Hughes, Rajinder Bhardwaj, Richard Bertz, Jing Liu
Zavegepant (ZAVZPRET™) is a high-affinity, selective, small-molecule calcitonin gene-related peptide receptor antagonist available for acute treatment of migraine in adults. A population pharmacokinetic analysis was performed to describe zavegepant plasma concentration-time course, characterize bioavailability, and identify covariates affecting zavegepant exposure. The model was developed and validated using data from 10 phase I clinical studies, wherein zavegepant was administered intravenously, intranasally, or orally to healthy adults and patients with migraine. Plasma concentration-time data were analyzed using nonlinear mixed-effects modeling. A three-compartment model with first-order elimination from the central compartment, and sequential zero- and first-order absorption best described the observed plasma concentration-time course of zavegepant. Bioavailability was 5.1% and 0.65% for intranasal and oral treatment, respectively; absorption rate constants were 5.8 and 0.8 h-1, respectively. Body weight-based empirical allometric scaling was applied using standard exponents (0.75 for clearance and 1 for volume of distribution). Age (range 18-71 years), race, ethnicity, sex, renal function, and co-administration of oral contraceptives or sumatriptan did not significantly change zavegepant pharmacokinetics. Moderate hepatic impairment (Child-Pugh score 7-9) or co-administration of rifampin decreased elimination clearance of oral zavegepant by ~40%. The zavegepant population pharmacokinetic model adequately characterized zavegepant concentration-time profiles, the bioavailability of intranasal and oral zavegepant, as well as the effect of intrinsic and extrinsic factors on zavegepant pharmacokinetics.
{"title":"Population pharmacokinetic modeling of zavegepant, a calcitonin gene-related peptide receptor antagonist, in healthy adults and patients with migraine.","authors":"Craig M Comisar, Jose Francis, Jim H Hughes, Rajinder Bhardwaj, Richard Bertz, Jing Liu","doi":"10.1002/psp4.13257","DOIUrl":"https://doi.org/10.1002/psp4.13257","url":null,"abstract":"<p><p>Zavegepant (ZAVZPRET™) is a high-affinity, selective, small-molecule calcitonin gene-related peptide receptor antagonist available for acute treatment of migraine in adults. A population pharmacokinetic analysis was performed to describe zavegepant plasma concentration-time course, characterize bioavailability, and identify covariates affecting zavegepant exposure. The model was developed and validated using data from 10 phase I clinical studies, wherein zavegepant was administered intravenously, intranasally, or orally to healthy adults and patients with migraine. Plasma concentration-time data were analyzed using nonlinear mixed-effects modeling. A three-compartment model with first-order elimination from the central compartment, and sequential zero- and first-order absorption best described the observed plasma concentration-time course of zavegepant. Bioavailability was 5.1% and 0.65% for intranasal and oral treatment, respectively; absorption rate constants were 5.8 and 0.8 h<sup>-1</sup>, respectively. Body weight-based empirical allometric scaling was applied using standard exponents (0.75 for clearance and 1 for volume of distribution). Age (range 18-71 years), race, ethnicity, sex, renal function, and co-administration of oral contraceptives or sumatriptan did not significantly change zavegepant pharmacokinetics. Moderate hepatic impairment (Child-Pugh score 7-9) or co-administration of rifampin decreased elimination clearance of oral zavegepant by ~40%. The zavegepant population pharmacokinetic model adequately characterized zavegepant concentration-time profiles, the bioavailability of intranasal and oral zavegepant, as well as the effect of intrinsic and extrinsic factors on zavegepant pharmacokinetics.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142568033","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jia Ning, Amita Pansari, Karen Rowland Yeo, Aki T. Heikkinen, Catriona Waitt, Lisa M. Almond
Optimal dosing in pregnant and lactating women requires an understanding of the pharmacokinetics in the mother, fetus, and breastfed infant. Physiologically-based pharmacokinetic (PBPK) modeling can be used to simulate untested scenarios and hence supplement clinical data to support dosing decisions. A PBPK model for the antiretroviral dolutegravir (mainly metabolized by UGT1A1) was verified using reported exposures in non-pregnant healthy volunteers, pregnant women, and the umbilical cord, lactating mothers, and breastfed neonates. The model was then applied to predict the impact of UGT1A1 phenotypes in extensive (EM), poor (PM), and ultra-rapid metabolizers (UM). The predicted dolutegravir maternal plasma and umbilical cord AUC in UGT1A1 PMs was 1.6-fold higher than in EMs. The predicted dolutegravir maternal plasma and umbilical cord AUC in UGT1A1 UMs mothers was 1.3-fold lower than in EMs. The predicted mean systemic and umbilical vein concentrations were in excess of the dolutegravir IC90 at 17, 28, and 40 gestational weeks, regardless of UGT1A1 phenotype, indicating that the standard dose of dolutegravir (50 mg q.d., fed state) is generally appropriate in late pregnancy, across UGT1A1 phenotypes. Applying the model in breastfed infants, a 1.5-, 1.7-, and 2.2-fold higher exposure in 2-day-old neonates, 10-day-old neonates, and infants who were UGT1A1 PMs, respectively, compared with EMs of the same age. However, it should be noted that the exposure in breastfed infants who were UGT1A1 PMs was still an order of magnitude lower than maternal exposure with a relative infant daily dose of <2%, suggesting safe use of dolutegravir in breastfeeding women.
{"title":"Using PBPK modeling to supplement clinical data and support the safe and effective use of dolutegravir in pregnant and lactating women","authors":"Jia Ning, Amita Pansari, Karen Rowland Yeo, Aki T. Heikkinen, Catriona Waitt, Lisa M. Almond","doi":"10.1002/psp4.13251","DOIUrl":"10.1002/psp4.13251","url":null,"abstract":"<p>Optimal dosing in pregnant and lactating women requires an understanding of the pharmacokinetics in the mother, fetus, and breastfed infant. Physiologically-based pharmacokinetic (PBPK) modeling can be used to simulate untested scenarios and hence supplement clinical data to support dosing decisions. A PBPK model for the antiretroviral dolutegravir (mainly metabolized by UGT1A1) was verified using reported exposures in non-pregnant healthy volunteers, pregnant women, and the umbilical cord, lactating mothers, and breastfed neonates. The model was then applied to predict the impact of UGT1A1 phenotypes in extensive (EM), poor (PM), and ultra-rapid metabolizers (UM). The predicted dolutegravir maternal plasma and umbilical cord AUC in UGT1A1 PMs was 1.6-fold higher than in EMs. The predicted dolutegravir maternal plasma and umbilical cord AUC in UGT1A1 UMs mothers was 1.3-fold lower than in EMs. The predicted mean systemic and umbilical vein concentrations were in excess of the dolutegravir IC<sub>90</sub> at 17, 28, and 40 gestational weeks, regardless of UGT1A1 phenotype, indicating that the standard dose of dolutegravir (50 mg q.d., fed state) is generally appropriate in late pregnancy, across UGT1A1 phenotypes. Applying the model in breastfed infants, a 1.5-, 1.7-, and 2.2-fold higher exposure in 2-day-old neonates, 10-day-old neonates, and infants who were UGT1A1 PMs, respectively, compared with EMs of the same age. However, it should be noted that the exposure in breastfed infants who were UGT1A1 PMs was still an order of magnitude lower than maternal exposure with a relative infant daily dose of <2%, suggesting safe use of dolutegravir in breastfeeding women.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"13 11","pages":"1924-1938"},"PeriodicalIF":3.1,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.13251","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142544234","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Byungwook Kim, Jung Eun Kim, Soyoung Lee, Jaeseong Oh, Joo-Youn Cho, In-Jin Jang, SeungHwan Lee, Jae-Yong Chung, Seonghae Yoon
Uremia, a condition characterized by the retention of uremic toxins due to impaired renal function, may affect drug metabolism mediated by CYP3A4 enzymes. Evogliptin is a dipeptidyl peptidase-4 (DPP-4) inhibitor diabetic drug that is primarily metabolized by CYP3A4. This study aimed to construct a population pharmacokinetic (PK) and pharmacodynamic (PD) model for evogliptin in patients with varying degrees of renal disease, including end-stage renal disease on hemodialysis. A total of 688 evogliptin concentration and 598 DPP-4 activity data were available from 46 subjects. PK and PD data analyses were performed using a nonlinear mixed-effects model. The PK of evogliptin was optimally described by a two-compartment model with first-order absorption. The significant covariates in the final model included blood amylase and triglyceride on F1 (relative bioavailability). The simulation findings, together with previously reported PK data, provided evidence of a significant inhibition of the first-pass effect of evogliptin in patients with renal impairment. A direct link sigmoidal Emax model was developed to describe the relationship between evogliptin concentration and DPP-4 inhibition. The PD model predicted significant inhibition of DPP-4 at maximum effect (Emax: 88.9%) and a low EC50 value (1.08 μg/L), indicating the high potency and efficacy of evogliptin. The developed PK/PD model accurately predicted exposure and the resulting DPP-4 activity of evogliptin in renal impairment. The findings of this study suggest that renal impairment and associated biochemical changes may impact the bioavailability of CYP3A4-metabolized drugs.
{"title":"Population pharmacokinetic and pharmacodynamic model of evogliptin: Severe uremia increases the bioavailability of evogliptin.","authors":"Byungwook Kim, Jung Eun Kim, Soyoung Lee, Jaeseong Oh, Joo-Youn Cho, In-Jin Jang, SeungHwan Lee, Jae-Yong Chung, Seonghae Yoon","doi":"10.1002/psp4.13263","DOIUrl":"https://doi.org/10.1002/psp4.13263","url":null,"abstract":"<p><p>Uremia, a condition characterized by the retention of uremic toxins due to impaired renal function, may affect drug metabolism mediated by CYP3A4 enzymes. Evogliptin is a dipeptidyl peptidase-4 (DPP-4) inhibitor diabetic drug that is primarily metabolized by CYP3A4. This study aimed to construct a population pharmacokinetic (PK) and pharmacodynamic (PD) model for evogliptin in patients with varying degrees of renal disease, including end-stage renal disease on hemodialysis. A total of 688 evogliptin concentration and 598 DPP-4 activity data were available from 46 subjects. PK and PD data analyses were performed using a nonlinear mixed-effects model. The PK of evogliptin was optimally described by a two-compartment model with first-order absorption. The significant covariates in the final model included blood amylase and triglyceride on F1 (relative bioavailability). The simulation findings, together with previously reported PK data, provided evidence of a significant inhibition of the first-pass effect of evogliptin in patients with renal impairment. A direct link sigmoidal E<sub>max</sub> model was developed to describe the relationship between evogliptin concentration and DPP-4 inhibition. The PD model predicted significant inhibition of DPP-4 at maximum effect (E<sub>max</sub>: 88.9%) and a low EC<sub>50</sub> value (1.08 μg/L), indicating the high potency and efficacy of evogliptin. The developed PK/PD model accurately predicted exposure and the resulting DPP-4 activity of evogliptin in renal impairment. The findings of this study suggest that renal impairment and associated biochemical changes may impact the bioavailability of CYP3A4-metabolized drugs.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142496549","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ahizechukwu C. Eke, Emily Adams, George U. Eleje, Ifeanyichukwu U. Ezebialu, Muktar H. Aliyu
<p>Although pharmacometric approaches play a critical role in modern drug development, their application in pregnancy is still limited, despite the widespread use of medications during gestation. Approximately 70%–80% of pregnant women use at least one prescription medication during the first trimester, and 90% take at least one medication during the course of their pregnancy<span><sup>2</sup></span>; yet, the effects of many of these drugs on pregnancy remain unknown. By leveraging complex mathematical models such as PBPK and PopPK approaches, researchers can simulate maternal and fetal drug exposure, optimize therapeutic regimens, and predict potential drug–drug interactions. The significant potential of pharmacometrics to address these critical issues in maternal and fetal pharmacology underscores the need for greater integration of these methodologies into clinical practice and research.</p><p>Pregnancy is a unique physiological state characterized by profound alterations in the absorption, distribution, metabolism, and elimination (ADME) of drugs.<span><sup>3</sup></span> Pregnancy-induced physiological changes affect multiple organ systems, including the cardiovascular, renal, hepatic, and gastrointestinal systems. As gestation progresses, maternal blood volume increases, glomerular filtration rate (GFR) rises, and hepatic enzyme activity is altered, impacting bioavailability, drug metabolism, and clearance.<span><sup>3</sup></span> For instance, in pregnancy, the activity of cytochrome P450 enzymes such as CYP3A4 increases while the activity of others like CYP1A2 decreases, leading to significantly greater variability in drug disposition.<span><sup>3</sup></span> These changes can pose significant challenges in determining optimal dosing, efficacy, and safety profiles for medications used during pregnancy, raising concern for both under- and overtreatment. Notably, most knowledge regarding the pharmacokinetics and safety of medications used during pregnancy is typically acquired 6–8 years after initial drug licensure,<span><sup>4</sup></span> highlighting the urgent need for advanced modeling approaches for earlier prediction of maternal and fetal drug exposure. Pharmacometrics provides an invaluable framework for addressing these challenges, making it indispensable in contemporary obstetrics and maternal–fetal-medicine research.</p><p>Pharmacometrics has shown utility in critical areas of obstetrics, particularly in predicting drug dosing and ensuring drug safety. For instance, PBPK models have effectively predicted maternal and fetal drug exposure for medications like nifedipine, allowing for safe management of preterm labor and pregnancy-induced hypertension.<span><sup>5</sup></span> Additionally, PopPK approaches have been employed to optimize dosing and to identify key covariates affecting drug disposition for magnesium sulfate administration for seizure prophylaxis in pre-eclampsia, considering factors such as altered plasma protein
{"title":"Pharmacometrics in obstetrics and maternal–fetal medicine research: Bridging gaps in maternal and fetal pharmacology","authors":"Ahizechukwu C. Eke, Emily Adams, George U. Eleje, Ifeanyichukwu U. Ezebialu, Muktar H. Aliyu","doi":"10.1002/psp4.13267","DOIUrl":"10.1002/psp4.13267","url":null,"abstract":"<p>Although pharmacometric approaches play a critical role in modern drug development, their application in pregnancy is still limited, despite the widespread use of medications during gestation. Approximately 70%–80% of pregnant women use at least one prescription medication during the first trimester, and 90% take at least one medication during the course of their pregnancy<span><sup>2</sup></span>; yet, the effects of many of these drugs on pregnancy remain unknown. By leveraging complex mathematical models such as PBPK and PopPK approaches, researchers can simulate maternal and fetal drug exposure, optimize therapeutic regimens, and predict potential drug–drug interactions. The significant potential of pharmacometrics to address these critical issues in maternal and fetal pharmacology underscores the need for greater integration of these methodologies into clinical practice and research.</p><p>Pregnancy is a unique physiological state characterized by profound alterations in the absorption, distribution, metabolism, and elimination (ADME) of drugs.<span><sup>3</sup></span> Pregnancy-induced physiological changes affect multiple organ systems, including the cardiovascular, renal, hepatic, and gastrointestinal systems. As gestation progresses, maternal blood volume increases, glomerular filtration rate (GFR) rises, and hepatic enzyme activity is altered, impacting bioavailability, drug metabolism, and clearance.<span><sup>3</sup></span> For instance, in pregnancy, the activity of cytochrome P450 enzymes such as CYP3A4 increases while the activity of others like CYP1A2 decreases, leading to significantly greater variability in drug disposition.<span><sup>3</sup></span> These changes can pose significant challenges in determining optimal dosing, efficacy, and safety profiles for medications used during pregnancy, raising concern for both under- and overtreatment. Notably, most knowledge regarding the pharmacokinetics and safety of medications used during pregnancy is typically acquired 6–8 years after initial drug licensure,<span><sup>4</sup></span> highlighting the urgent need for advanced modeling approaches for earlier prediction of maternal and fetal drug exposure. Pharmacometrics provides an invaluable framework for addressing these challenges, making it indispensable in contemporary obstetrics and maternal–fetal-medicine research.</p><p>Pharmacometrics has shown utility in critical areas of obstetrics, particularly in predicting drug dosing and ensuring drug safety. For instance, PBPK models have effectively predicted maternal and fetal drug exposure for medications like nifedipine, allowing for safe management of preterm labor and pregnancy-induced hypertension.<span><sup>5</sup></span> Additionally, PopPK approaches have been employed to optimize dosing and to identify key covariates affecting drug disposition for magnesium sulfate administration for seizure prophylaxis in pre-eclampsia, considering factors such as altered plasma protein ","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"13 11","pages":"1835-1840"},"PeriodicalIF":3.1,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.13267","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142496548","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Stan is a powerful probabilistic programming language designed mainly for Bayesian data analysis. Torsten is a collection of Stan functions that handles the events (e.g., dosing events) and solves the ODE systems that are frequently present in pharmacometric models. To perform a Bayesian data analysis, most models in pharmacometrics require Markov Chain Monte Carlo (MCMC) methods to sample from the posterior distribution. However, MCMC is computationally expensive and can be time-consuming, enough so that people will often forgo Bayesian methods for a more traditional approach. This paper shows how to speed up the sampling process in Stan by within-chain parallelization through both multi-threading using Stan's reduce_sum() function and multi-processing using Torsten's group ODE solver. Both methods show substantial reductions in the time necessary to sufficiently sample from the posterior distribution compared with a basic approach with no within-chain parallelization.
Stan 是一种功能强大的概率编程语言,主要用于贝叶斯数据分析。Torsten 是一组 Stan 函数,用于处理事件(如用药事件)和解决药物计量学模型中经常出现的 ODE 系统。要进行贝叶斯数据分析,药物计量学中的大多数模型都需要用马尔可夫链蒙特卡罗(MCMC)方法从后验分布中采样。然而,MCMC 的计算成本很高,而且非常耗时,因此人们往往会放弃贝叶斯方法,转而采用更传统的方法。本文展示了如何通过使用 Stan 的 reduce_sum() 函数进行多线程处理和使用 Torsten 的组 ODE 求解器进行多进程处理,在 Stan 中通过链内并行化加速采样过程。与没有链内并行化的基本方法相比,这两种方法都显示出从后验分布中充分采样所需的时间大幅减少。
{"title":"Within-chain parallelization-Giving Stan Jet Fuel for population modeling in pharmacometrics.","authors":"Casey Davis, Pavan Vaddady","doi":"10.1002/psp4.13238","DOIUrl":"https://doi.org/10.1002/psp4.13238","url":null,"abstract":"<p><p>Stan is a powerful probabilistic programming language designed mainly for Bayesian data analysis. Torsten is a collection of Stan functions that handles the events (e.g., dosing events) and solves the ODE systems that are frequently present in pharmacometric models. To perform a Bayesian data analysis, most models in pharmacometrics require Markov Chain Monte Carlo (MCMC) methods to sample from the posterior distribution. However, MCMC is computationally expensive and can be time-consuming, enough so that people will often forgo Bayesian methods for a more traditional approach. This paper shows how to speed up the sampling process in Stan by within-chain parallelization through both multi-threading using Stan's reduce_sum() function and multi-processing using Torsten's group ODE solver. Both methods show substantial reductions in the time necessary to sufficiently sample from the posterior distribution compared with a basic approach with no within-chain parallelization.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142496550","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Breastfeeding is important in childhood development, and medications are often necessary for lactating individuals, yet information on the potential risk of infant drug exposure through human milk is limited. Establishing a lactation modeling framework can advance our understanding of this topic and potentiate clinical decision making. We expanded the modeling framework previously developed for sotalol using pregabalin as a second prototypical probe compound with similar absorption, distribution, metabolism, and elimination (ADME) properties. Adult oral models were developed in PK-Sim® and used to build a lactation model in MoBi® to simulate drug transfer into human milk. The adult model was applied to breastfeeding pediatrics (ages 1 to 23 months) and subsequently integrated with the lactation model to simulate infant drug exposure according to age, size, and breastfeeding frequency. Physiologically based pharmacokinetic (PBPK) model simulations captured the data used for verification both in adults and pediatrics. Lactation simulations captured observed milk and plasma data corresponding to doses of 150 mg administered twice daily to lactating individuals, and estimated a relative infant dose (RID) of approximately 7% of the maternal dose. The infant drug exposure simulations showed peak plasma concentrations of 0.44 μg/mL occurring within the first 2 weeks of life, followed by gradual decline with age after week four. The modeling framework performs well for this second prototypical drug and warrants expansion to other drugs for further validation. PBPK modeling and simulation approaches together with clinical lactation data could ultimately help inform infant drug exposure risk assessments to guide clinical decision making.
{"title":"Informing the risk assessment related to lactation and drug exposure: A physiologically based pharmacokinetic lactation model for pregabalin","authors":"Cameron Humerickhouse, Michelle Pressly, Zhoumeng Lin, Daphne Guinn, Sherbet Samuels, Elimika Pfuma Fletcher, Stephan Schmidt","doi":"10.1002/psp4.13266","DOIUrl":"10.1002/psp4.13266","url":null,"abstract":"<p>Breastfeeding is important in childhood development, and medications are often necessary for lactating individuals, yet information on the potential risk of infant drug exposure through human milk is limited. Establishing a lactation modeling framework can advance our understanding of this topic and potentiate clinical decision making. We expanded the modeling framework previously developed for sotalol using pregabalin as a second prototypical probe compound with similar absorption, distribution, metabolism, and elimination (ADME) properties. Adult oral models were developed in PK-Sim® and used to build a lactation model in MoBi® to simulate drug transfer into human milk. The adult model was applied to breastfeeding pediatrics (ages 1 to 23 months) and subsequently integrated with the lactation model to simulate infant drug exposure according to age, size, and breastfeeding frequency. Physiologically based pharmacokinetic (PBPK) model simulations captured the data used for verification both in adults and pediatrics. Lactation simulations captured observed milk and plasma data corresponding to doses of 150 mg administered twice daily to lactating individuals, and estimated a relative infant dose (RID) of approximately 7% of the maternal dose. The infant drug exposure simulations showed peak plasma concentrations of 0.44 μg/mL occurring within the first 2 weeks of life, followed by gradual decline with age after week four. The modeling framework performs well for this second prototypical drug and warrants expansion to other drugs for further validation. PBPK modeling and simulation approaches together with clinical lactation data could ultimately help inform infant drug exposure risk assessments to guide clinical decision making.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"13 11","pages":"1953-1966"},"PeriodicalIF":3.1,"publicationDate":"2024-10-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.13266","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142496546","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Christian Bartels, Martina Scauda, Neva Coello, Thomas Dumortier, Björn Bornkamp, Giusi Moffa
The ICH E9 (R1) guidance and the related estimand framework propose to clearly define and separate the clinical question of interest formulated as estimand from the estimation method. With that it becomes important to assess the validity of the estimation method and the assumptions that must be made. When going beyond the intention to treat analyses that can rely on randomization, causal inference is usually used to discuss the validity of estimation methods for the estimand of interest. In pharmacometrics, mixed-effects models are routinely used to analyze longitudinal clinical trial data; however, they are rarely discussed as a method for causal inference. Here, we evaluate nonlinear mixed-effects modeling and simulation (NLME M&S) in the context of causal inference as a standardization method for longitudinal data in the presence of confounders. Standardization is a well-known method in causal inference to correct for confounding by analyzing and combining results from subgroups of patients. We show that nonlinear mixed-effects modeling is a particular implementation of standardization that conditions on individual parameters described by the random effects of the mixed-effects model. As an example, we use a simulated clinical trial with within-subject dose titration. Being interested in the outcome of the hypothetical situation that patients adhere to the planned treatment schedule, we put assumptions in a causal diagram. From the causal diagram, conditional independence assumptions are derived either by conditioning on the individual parameters or on earlier outcomes. With both conditional independencies unbiased estimates can be obtained.
ICH E9 (R1)指南和相关的估计值框架建议明确定义作为估计值的临床相关问题,并将其与估计方法分开。因此,评估估算方法的有效性和必须做出的假设就变得非常重要。在超越可以依赖随机化的意向治疗分析时,因果推论通常被用来讨论估计方法对所关注估计对象的有效性。在药物计量学中,混合效应模型通常用于分析纵向临床试验数据;然而,它们很少被作为因果推断的一种方法来讨论。在此,我们评估了非线性混合效应建模和模拟(NLME M&S)在因果推断中作为存在混杂因素的纵向数据标准化方法的应用情况。标准化是因果推断中一种众所周知的方法,通过分析和合并来自亚组患者的结果来校正混杂因素。我们表明,非线性混合效应模型是标准化的一种特殊实现方式,它以混合效应模型随机效应所描述的单个参数为条件。举例来说,我们使用了一个具有受试者内剂量滴定功能的模拟临床试验。我们对患者按计划接受治疗这一假设情况的结果感兴趣,因此在因果图中加入了假设条件。从因果图中,通过对单个参数或早期结果进行条件限制,得出条件独立性假设。有了这两种条件独立性,就可以得到无偏估计值。
{"title":"Nonlinear mixed-effects modeling as a method for causal inference to predict exposures under desired within-subject dose titration schemes.","authors":"Christian Bartels, Martina Scauda, Neva Coello, Thomas Dumortier, Björn Bornkamp, Giusi Moffa","doi":"10.1002/psp4.13239","DOIUrl":"https://doi.org/10.1002/psp4.13239","url":null,"abstract":"<p><p>The ICH E9 (R1) guidance and the related estimand framework propose to clearly define and separate the clinical question of interest formulated as estimand from the estimation method. With that it becomes important to assess the validity of the estimation method and the assumptions that must be made. When going beyond the intention to treat analyses that can rely on randomization, causal inference is usually used to discuss the validity of estimation methods for the estimand of interest. In pharmacometrics, mixed-effects models are routinely used to analyze longitudinal clinical trial data; however, they are rarely discussed as a method for causal inference. Here, we evaluate nonlinear mixed-effects modeling and simulation (NLME M&S) in the context of causal inference as a standardization method for longitudinal data in the presence of confounders. Standardization is a well-known method in causal inference to correct for confounding by analyzing and combining results from subgroups of patients. We show that nonlinear mixed-effects modeling is a particular implementation of standardization that conditions on individual parameters described by the random effects of the mixed-effects model. As an example, we use a simulated clinical trial with within-subject dose titration. Being interested in the outcome of the hypothetical situation that patients adhere to the planned treatment schedule, we put assumptions in a causal diagram. From the causal diagram, conditional independence assumptions are derived either by conditioning on the individual parameters or on earlier outcomes. With both conditional independencies unbiased estimates can be obtained.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142496547","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Nihan Izat, Jayaprakasam Bolleddula, Pasquale Carione, Leticia Huertas Valentin, Robert S Jones, Priyanka Kulkarni, Darren Moss, Vincent C Peterkin, Dan-Dan Tian, Andrea Treyer, Karthik Venkatakrishnan, Michael A Zientek, Jill Barber, J Brian Houston, Aleksandra Galetin, Daniel Scotcher
Aldehyde oxidase (AO) contributes to the clearance of many approved and investigational small molecule drugs, which are often dual substrates of AO and drug-metabolizing enzymes such as cytochrome P450s (CYPs). As such, the lack of established framework for quantitative translation of the clinical pharmacologic correlates of AO-mediated clearance represents an unmet need. This study aimed to evaluate the utility of physiologically based pharmacokinetic (PBPK) modeling in the development of AO and dual AO-CYP substrates. PBPK models were developed for capmatinib, idelalisib, lenvatinib, zaleplon, ziprasidone, and zoniporide, incorporating in vitro functional data from human liver subcellular fractions and human hepatocytes. Prediction of metabolic elimination with/without the additional empirical scaling factors (ESFs) was assessed. Clinical pharmacokinetics, human mass balance, and drug-drug interaction (DDI) studies with CYP3A4 modulators, where available, were used to refine/verify the models. Due to the lack of clinically significant AO-DDIs with known AO inhibitors, the fraction metabolized by AO (fmAO) was verified indirectly. Clearance predictions were improved by using ESFs (GMFE ≤1.4-fold versus up to fivefold with physiologically-based scaling only). Observed fmi from mass balance studies were crucial for model verification/refinement, as illustrated by capmatinib, where the fmAO (40%) was otherwise underpredicted up to fourfold. Subsequently, independent DDI studies with ketoconazole, itraconazole, rifampicin, and carbamazepine verified the fmCYP3A4, with predicted ratios of the area under the concentration-time curve (AUCR) within 1.5-fold of the observations. In conclusion, this study provides a novel PBPK-based framework for predicting AO-mediated pharmacokinetics and quantitative assessment of clinical DDI risks for dual AO-CYP substrates within a totality-of-evidence approach.
醛氧化酶(AO)有助于清除许多已批准和在研的小分子药物,这些药物通常是 AO 和药物代谢酶(如细胞色素 P450s,CYPs)的双重底物。因此,缺乏对 AO 介导的清除率的临床药理学相关性进行定量转化的既定框架是一项尚未满足的需求。本研究旨在评估基于生理学的药代动力学(PBPK)模型在 AO 和 AO-CYP 双底物开发中的实用性。结合人肝亚细胞组分和人肝细胞的体外功能数据,为卡马替尼、伊德拉利西、来伐替尼、扎来普隆、齐拉西酮和佐尼波利开发了PBPK模型。评估了使用/不使用附加经验缩放因子(ESF)的代谢消除预测。临床药代动力学、人体质量平衡以及与 CYP3A4 调节剂的药物相互作用 (DDI) 研究(如有)被用来完善/验证模型。由于缺乏与已知 AO 抑制剂的具有临床意义的 AO-DDI 研究,因此间接验证了经 AO 代谢的部分(fmAO)。通过使用 ESF,清除率预测得到了改善(GMFE ≤ 1.4 倍,而仅使用基于生理学的缩放比例则高达 5 倍)。质量平衡研究中观察到的 fmi 对模型验证/改进至关重要,卡马替尼就是一例,其 fmAO(40%)被低估了四倍。随后,对酮康唑、伊曲康唑、利福平和卡马西平进行的独立 DDI 研究验证了 fmCYP3A4,预测的浓度-时间曲线下面积(AUCR)比值在观察值的 1.5 倍以内。总之,本研究提供了一种基于 PBPK 的新框架,用于预测 AO 介导的药代动力学,并以证据整体法定量评估 AO-CYP 双底物的临床 DDI 风险。
{"title":"Establishing a physiologically based pharmacokinetic framework for aldehyde oxidase and dual aldehyde oxidase-CYP substrates.","authors":"Nihan Izat, Jayaprakasam Bolleddula, Pasquale Carione, Leticia Huertas Valentin, Robert S Jones, Priyanka Kulkarni, Darren Moss, Vincent C Peterkin, Dan-Dan Tian, Andrea Treyer, Karthik Venkatakrishnan, Michael A Zientek, Jill Barber, J Brian Houston, Aleksandra Galetin, Daniel Scotcher","doi":"10.1002/psp4.13255","DOIUrl":"https://doi.org/10.1002/psp4.13255","url":null,"abstract":"<p><p>Aldehyde oxidase (AO) contributes to the clearance of many approved and investigational small molecule drugs, which are often dual substrates of AO and drug-metabolizing enzymes such as cytochrome P450s (CYPs). As such, the lack of established framework for quantitative translation of the clinical pharmacologic correlates of AO-mediated clearance represents an unmet need. This study aimed to evaluate the utility of physiologically based pharmacokinetic (PBPK) modeling in the development of AO and dual AO-CYP substrates. PBPK models were developed for capmatinib, idelalisib, lenvatinib, zaleplon, ziprasidone, and zoniporide, incorporating in vitro functional data from human liver subcellular fractions and human hepatocytes. Prediction of metabolic elimination with/without the additional empirical scaling factors (ESFs) was assessed. Clinical pharmacokinetics, human mass balance, and drug-drug interaction (DDI) studies with CYP3A4 modulators, where available, were used to refine/verify the models. Due to the lack of clinically significant AO-DDIs with known AO inhibitors, the fraction metabolized by AO (fm<sub>AO</sub>) was verified indirectly. Clearance predictions were improved by using ESFs (GMFE ≤1.4-fold versus up to fivefold with physiologically-based scaling only). Observed fm<sub>i</sub> from mass balance studies were crucial for model verification/refinement, as illustrated by capmatinib, where the fm<sub>AO</sub> (40%) was otherwise underpredicted up to fourfold. Subsequently, independent DDI studies with ketoconazole, itraconazole, rifampicin, and carbamazepine verified the fm<sub>CYP3A4</sub>, with predicted ratios of the area under the concentration-time curve (AUCR) within 1.5-fold of the observations. In conclusion, this study provides a novel PBPK-based framework for predicting AO-mediated pharmacokinetics and quantitative assessment of clinical DDI risks for dual AO-CYP substrates within a totality-of-evidence approach.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-10-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142496545","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jane P F Bai, Guansheng Liu, Miao Zhao, Jie Wang, Ye Xiong, Tien Truong, Justin C Earp, Yuching Yang, Jiang Liu, Hao Zhu, Gilbert J Burckart
The number of quantitative systems pharmacology (QSP) submissions to the U.S. Food and Drug Administration has continued to increase over the past decade. This report summarizes the landscape of QSP submissions as of December 2023. QSP was used to inform drug development across various therapeutic areas and throughout the drug development process of small molecular drugs and biologics and has facilitated dose finding, dose ranging, and dose optimization studies. Though the majority of QSP submissions (>66%) focused on drug effectiveness, QSP was also utilized to simulate drug safety including liver toxicity, risk of cytokine release syndrome (CRS), bone density, and others. This report also includes individual contexts of use from a handful of new drug applications (NDAs) and biologics license applications where QSP modeling was used to demonstrate the utility of QSP modeling in regulatory drug development. According to the models submitted in QSP submissions, an anonymous case was utilized to illustrate how QSP informed development of a bispecific monoclonal antibody with respect to CRS risk. QSP submissions for informing pediatric drug development were summarized along with highlights of a case in inborn errors of metabolism. Furthermore, simulations of response variability with QSP were described. In summary, QSP continues to play a role in informing drug development.
{"title":"Landscape of regulatory quantitative systems pharmacology submissions to the U.S. Food and Drug Administration: An update report.","authors":"Jane P F Bai, Guansheng Liu, Miao Zhao, Jie Wang, Ye Xiong, Tien Truong, Justin C Earp, Yuching Yang, Jiang Liu, Hao Zhu, Gilbert J Burckart","doi":"10.1002/psp4.13208","DOIUrl":"https://doi.org/10.1002/psp4.13208","url":null,"abstract":"<p><p>The number of quantitative systems pharmacology (QSP) submissions to the U.S. Food and Drug Administration has continued to increase over the past decade. This report summarizes the landscape of QSP submissions as of December 2023. QSP was used to inform drug development across various therapeutic areas and throughout the drug development process of small molecular drugs and biologics and has facilitated dose finding, dose ranging, and dose optimization studies. Though the majority of QSP submissions (>66%) focused on drug effectiveness, QSP was also utilized to simulate drug safety including liver toxicity, risk of cytokine release syndrome (CRS), bone density, and others. This report also includes individual contexts of use from a handful of new drug applications (NDAs) and biologics license applications where QSP modeling was used to demonstrate the utility of QSP modeling in regulatory drug development. According to the models submitted in QSP submissions, an anonymous case was utilized to illustrate how QSP informed development of a bispecific monoclonal antibody with respect to CRS risk. QSP submissions for informing pediatric drug development were summarized along with highlights of a case in inborn errors of metabolism. Furthermore, simulations of response variability with QSP were described. In summary, QSP continues to play a role in informing drug development.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142459959","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}