Journal of Pharmacokinetics and Pharmacodynamics最新文献

筛选
英文 中文
Novel endpoints based on tumor size ratio to support early clinical decision-making in oncology drug-development.
IF 2.2 4区 医学
Journal of Pharmacokinetics and Pharmacodynamics Pub Date : 2024-12-20 DOI: 10.1007/s10928-024-09946-3
Shubhadeep Chakraborty, Kshitij Aggarwal, Marzana Chowdhury, Izumi Hamada, Chuanpu Hu, Anna Kondic, Kaushal Mishra, David Paulucci, Ram Tiwari, Kalyanee Viraswami Appanna, Mariann Micsinai Balan, Arun Kumar
{"title":"Novel endpoints based on tumor size ratio to support early clinical decision-making in oncology drug-development.","authors":"Shubhadeep Chakraborty, Kshitij Aggarwal, Marzana Chowdhury, Izumi Hamada, Chuanpu Hu, Anna Kondic, Kaushal Mishra, David Paulucci, Ram Tiwari, Kalyanee Viraswami Appanna, Mariann Micsinai Balan, Arun Kumar","doi":"10.1007/s10928-024-09946-3","DOIUrl":"https://doi.org/10.1007/s10928-024-09946-3","url":null,"abstract":"<p><p>In oncology drug development, overall response rate (ORR) is commonly used as an early endpoint to assess the clinical benefits of new interventions; however, ORR benefit may not always translate into a long-term clinical benefit such as overall survival (OS). Most of the work on developing endpoints based on tumor growth dynamics relies on empirical validation, leading to a lack of generalizability of the endpoints across indications and therapeutic modalities. Additionally, many of these metrics are model-based and do not use data from all the patients. The objective of this work is to use longitudinal tumor size data and new lesion information (that is, the same information used by the ORR) to develop novel endpoints that can improve early clinical decision-making compared to the ORR. We investigate in this work multiple candidate novel endpoints based on tumor size ratio that utilize longitudinal tumor size data from all the patients regardless of their follow-up, rely only on tumor size and new lesion information, and are model-free. An extensive simulation study is conducted, exploring a wide spectrum of tumor size data and overall survival outcomes by modulating a variety of trial characteristics such as slow vs fast tumor growth, high vs low drug efficacy rates, variability in patients' responses, variations in the number of patients, follow-up periods, new lesion rates and survival curve shapes. The proposed novel endpoints based on tumor size ratio consistently outperform the ORR by having a comparable or higher correlation with the OS. Further, the novel endpoints exhibit superior accuracy compared to the ORR in predicting the long-term OS benefit. Retrospective empirical validation on BMS clinical trials confirms our simulation findings. These findings suggest that the tumor size ratio-based endpoints could replace ORR for early clinical decision-making in oncology drug development.</p>","PeriodicalId":16851,"journal":{"name":"Journal of Pharmacokinetics and Pharmacodynamics","volume":"52 1","pages":"9"},"PeriodicalIF":2.2,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142864635","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Translational pharmacokinetic and pharmacodynamic modelling of the anti-ADAMTS-5 NANOBODY® (M6495) using the neo-epitope ARGS as a biomarker.
IF 2.2 4区 医学
Journal of Pharmacokinetics and Pharmacodynamics Pub Date : 2024-12-20 DOI: 10.1007/s10928-024-09958-z
Joao N S Pereira, Ingrid Ottevaere, Benedikte Serruys, Hans Guehring, Christoph Ladel, Sven Lindemann
{"title":"Translational pharmacokinetic and pharmacodynamic modelling of the anti-ADAMTS-5 NANOBODY<sup>®</sup> (M6495) using the neo-epitope ARGS as a biomarker.","authors":"Joao N S Pereira, Ingrid Ottevaere, Benedikte Serruys, Hans Guehring, Christoph Ladel, Sven Lindemann","doi":"10.1007/s10928-024-09958-z","DOIUrl":"https://doi.org/10.1007/s10928-024-09958-z","url":null,"abstract":"<p><p>M6495 is a first-in-class NANOBODY<sup>®</sup> molecule and an inhibitor of ADAMTS-5, with the potential to be a disease modifying osteoarthritis drug. In order to investigate the PK/PD (pharmacokinetic and pharmacodynamic) properties of M6495, a single dose study was performed in cynomolgus monkeys with doses up to 6 mg/kg, with the goal of understanding the PK/PD properties of M6495. The neo-epitope ARGS (Alanine-Arginine-Glycine-Serine) generated by cleavage of aggrecan by ADAMTS-5 was used as a target-engagement biomarker. A long-lasting dose-dependent decrease in serum ARGS could be observed after a single dose of M6495 in cynomolgus monkeys. The serum biomarker ARGS decreased to levels below the limit of quantification of the assay in animals which received doses of M6495 of 6 mg/kg and higher, indicating a strong inhibition of ADAMTS-5. Data from the single-dose PK/PD study was combined with data from a multiple dose study, and a non-linear mixed effects model was used to explore the relationship between plasma concentrations of M6495 and the reduction of serum ARGS. The model was subsequently used to inform the clinical phase 1 study design and was successful in predicting the human clinical pharmacokinetics and pharmacodynamics of M6495. In addition to having enabled a Phase 1 trial with M6495, this is the first PK/PD model describing the pharmacodynamics of the neo-epitope ARGS after ADAMTS5 inhibition. It is expected that in the future, this model can be used or adapted to explore the PK/PD relationship between M6495 serum concentrations and the ARGS serum biomarker.</p>","PeriodicalId":16851,"journal":{"name":"Journal of Pharmacokinetics and Pharmacodynamics","volume":"52 1","pages":"8"},"PeriodicalIF":2.2,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142864641","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
QSP modeling of a transiently inactivating antibody-drug conjugate highlights benefit of short antibody half life.
IF 2.2 4区 医学
Journal of Pharmacokinetics and Pharmacodynamics Pub Date : 2024-12-17 DOI: 10.1007/s10928-024-09956-1
Eshita Khera, Lekshmi Dharmarajan, Dominik Hainzl, Volker Engelhardt, Helena Vostiarova, John Davis, Nicolas Ebel, Kuno Wuersch, Vincent Romanet, Sherif Sharaby, Jeffrey D Kearns
{"title":"QSP modeling of a transiently inactivating antibody-drug conjugate highlights benefit of short antibody half life.","authors":"Eshita Khera, Lekshmi Dharmarajan, Dominik Hainzl, Volker Engelhardt, Helena Vostiarova, John Davis, Nicolas Ebel, Kuno Wuersch, Vincent Romanet, Sherif Sharaby, Jeffrey D Kearns","doi":"10.1007/s10928-024-09956-1","DOIUrl":"https://doi.org/10.1007/s10928-024-09956-1","url":null,"abstract":"<p><p>Antibody drug conjugates (ADC) are a promising class of oncology therapeutics consisting of an antibody conjugated to a payload via a linker. DYP688 is a novel ADC comprising of a signaling protein inhibitor payload (FR900359) that undergoes unique on-antibody inactivation in plasma, resulting in complex pharmacology. To assess the impact of FR inactivation on DYP688 pharmacology and clinical developability, we performed translational modeling of preclinical PK and tumor growth inhibition (TGI) data, accompanied by mechanistic Krogh cylinder tumor modeling. Using a PK-TGI model, we identified a composite exposure-above-tumorostatic concentration (AUC<sub>TSC</sub>) metric as the PK-driver of efficacy. To underpin the mechanisms behind AUC<sub>TSC</sub> as the driver of efficacy, we performed quantitative systems pharmacology (QSP) modeling of DYP688 intratumoral pharmacokinetics and pharmacodynamics. Through exploratory simulations, we show that by deviating from canonical ADC design dogma, DYP688 has optimal FR900359 activity despite its transient inactivation. Finally, we performed the successful preclinical to clinical translation of DYP688 PK, including the payload inactivation kinetics, evidenced by good agreement of the predicted PK to the observed interim clinical PK. Overall, this work highlights early quantitative pharmacokinetics as a missing link in the ADC design-developability chasm.</p>","PeriodicalId":16851,"journal":{"name":"Journal of Pharmacokinetics and Pharmacodynamics","volume":"52 1","pages":"7"},"PeriodicalIF":2.2,"publicationDate":"2024-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142846746","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A PopPBPK-RL approach for precision dosing of benazepril in renal impaired patients.
IF 2.2 4区 医学
Journal of Pharmacokinetics and Pharmacodynamics Pub Date : 2024-12-11 DOI: 10.1007/s10928-024-09953-4
Guillermo Vigueras, Lucía Muñoz-Gil, Valerie Reinisch, Joana T Pinto
{"title":"A PopPBPK-RL approach for precision dosing of benazepril in renal impaired patients.","authors":"Guillermo Vigueras, Lucía Muñoz-Gil, Valerie Reinisch, Joana T Pinto","doi":"10.1007/s10928-024-09953-4","DOIUrl":"https://doi.org/10.1007/s10928-024-09953-4","url":null,"abstract":"<p><p>Current treatment recommendations mainly rely on rule-based protocols defined from evidence-based clinical guidelines, which are difficult to adapt for high-risk patients such as those with renal impairment. Consequently, unsuccessful therapies and the occurrence of adverse drug reactions are common. Within the context of personalized medicine, that tries to deliver the right treatment dose to maximize efficacy and minimize toxicity, the concept of model-informed precision dosing proposes the use of mechanistic models, like physiologically based pharmacokinetic (PBPK) modeling, to predict drug regimes outcomes. Nonetheless, PBPK models have limited capability when computing patients' centric optimized drug doses. Consequently, reinforcement learning (RL) has been previously used to personalize drug dosage. In this work we propose the first PBPK and RL-based precision dosing system for an orally taken drug (benazepril) considering a virtual population of patients with renal disease. Population based PBPK modeling is used in combination with RL for obtaining patient tailored dose regimes. We also perform patient stratification and feature selection to better handle dose tailoring problems. Based on patients' characteristics with best predictive capabilities, benazepril dose regimes are obtained for a population with features' diversity. Obtained regimes are evaluated based on PK parameters considered. Results show that the proof-of-concept approach herein is capable of learning good dosing regimes for most patients. The use of a PopPBPK model allowed to account for intervariability of patient characteristics and be more inclusive considering also non-frequent patients. Impact analysis of patients' features reveals that renal impairment is the main driver affecting RL capabilities.</p>","PeriodicalId":16851,"journal":{"name":"Journal of Pharmacokinetics and Pharmacodynamics","volume":"52 1","pages":"6"},"PeriodicalIF":2.2,"publicationDate":"2024-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142813535","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Comparison of the power and type 1 error of total score models for drug effect detection in clinical trials.
IF 2.2 4区 医学
Journal of Pharmacokinetics and Pharmacodynamics Pub Date : 2024-12-10 DOI: 10.1007/s10928-024-09949-0
Elham Haem, Mats O Karlsson, Sebastian Ueckert
{"title":"Comparison of the power and type 1 error of total score models for drug effect detection in clinical trials.","authors":"Elham Haem, Mats O Karlsson, Sebastian Ueckert","doi":"10.1007/s10928-024-09949-0","DOIUrl":"10.1007/s10928-024-09949-0","url":null,"abstract":"<p><p>Composite scale data consists of numerous categorical questions/items that are often summed as a total score and are commonly utilized as primary endpoints in clinical trials. These endpoints are conceptually discrete and constrained by nature. Item response theory (IRT) is a powerful approach for detecting drug effects in composite scale data from clinical trials, but estimating all parameters requires a large sample size and all item information, which may not be available. Therefore, total score models are often utilized. The most popular total score models are continuous variable (CV) models, but this strategy establishes assumptions that go against the integer nature, and typically also the bounded nature, of data. Bounded integer (BI) and Coarsened grid (CG) models respect the nature of the data. However, their power to detect drug effects has not been as thoroughly studied in clinical trials. When an IRT model is accessible, IRT-informed models (I-BI and I-CV) are promising methods in which the mean and variability of the total score at any position are extracted from the existing IRT model. In this study, total score data were simulated from the MDS-UPDRS motor subscale. Then, the power, type 1 error, and treatment effect bias of six total score models for detecting drug effects in clinical trials were explored. Further, it was investigated how the power, type 1 of error, and treatment effect bias for the I-BI and I-CV models were affected by mis-specified item information from the IRT model. The I-BI model demonstrated the highest statistical power, maintained an acceptable Type I error rate, and exhibited minimal bias, approaching zero. Following that, the I-CV, BI, and CG with Czado transformation (CG_Czado) models provided the maximum power. However, the CG_Czado model had inflated type 1 error under low sample size scenarios in each arm of clinical trials. The CG model among total score models displayed the lowest power and the most inflated type 1 error. Therefore, the results favor the I-BI model when an IRT model is available; otherwise, the BI model.</p>","PeriodicalId":16851,"journal":{"name":"Journal of Pharmacokinetics and Pharmacodynamics","volume":"52 1","pages":"4"},"PeriodicalIF":2.2,"publicationDate":"2024-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11632077/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142801232","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Model-informed approach to estimate treatment effect in placebo-controlled clinical trials using an artificial intelligence-based propensity weighting methodology to account for non-specific responses to treatment.
IF 2.2 4区 医学
Journal of Pharmacokinetics and Pharmacodynamics Pub Date : 2024-12-10 DOI: 10.1007/s10928-024-09950-7
Roberto Gomeni, F Bressolle-Gomeni
{"title":"Model-informed approach to estimate treatment effect in placebo-controlled clinical trials using an artificial intelligence-based propensity weighting methodology to account for non-specific responses to treatment.","authors":"Roberto Gomeni, F Bressolle-Gomeni","doi":"10.1007/s10928-024-09950-7","DOIUrl":"10.1007/s10928-024-09950-7","url":null,"abstract":"<p><p>In randomized, placebo controlled clinical trials (RCT) in major depressive disorders (MDD), treatment response (TR) is estimated by the change from baseline at study-end (EOS) of the scores of clinical scales used for assessing disease severity. Treatment effect (TE) is estimated by the baseline-adjusted difference at EOS of TR between active treatments and placebo.The TE is function of treatment-specific and, non-specific (NSRT) effect (referred as placebo effect), and placebo response. The conventional statistical approaches used to estimate TE does not account for the potentially confounding effect of NSRT. This pragmatic approach is equivalent to assume that TE is independent of NSRT even if this assumption is not true, leading to potential risks of inflating false negative/positive results in presence of high proportion of subjects with high/low NSRT.The objective of this study was to develop a model informed framework to analyze the outcomes of RCTs using data driven models, non-linear-mixed effect approach, artificial intelligence, and propensity score weighted methodology (PSW) to control the confounding effect of treatment non-specific response on the estimated TE. The secondary objective was to explore the impact of relevant covariates (including the assessment of a dose-response relationship) on the outcomes of pooled data from two RCTs.The proposed PSW approach provides a critical tool for controlling the confounding effect of treatment non-specific response, to increase signal detection and to provide a reliable estimate of the 'true' treatment effect by controlling false negative results associated with excessively high treatment non-specific response.</p>","PeriodicalId":16851,"journal":{"name":"Journal of Pharmacokinetics and Pharmacodynamics","volume":"52 1","pages":"5"},"PeriodicalIF":2.2,"publicationDate":"2024-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11631816/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142801239","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Translational population target binding model for the anti-FcRn fragment antibody efgartigimod.
IF 2.2 4区 医学
Journal of Pharmacokinetics and Pharmacodynamics Pub Date : 2024-12-05 DOI: 10.1007/s10928-024-09952-5
Sven Hoefman, Tamara van Steeg, Ingrid Ottevaere, Judith Baumeister, Stefaan Rossenu
{"title":"Translational population target binding model for the anti-FcRn fragment antibody efgartigimod.","authors":"Sven Hoefman, Tamara van Steeg, Ingrid Ottevaere, Judith Baumeister, Stefaan Rossenu","doi":"10.1007/s10928-024-09952-5","DOIUrl":"10.1007/s10928-024-09952-5","url":null,"abstract":"<p><p>Efgartigimod is a human IgG1 antibody Fc-fragment that lowers IgG levels through blockade of the neonatal Fc receptor (FcRn) and is being evaluated for the treatment of patients with severe autoimmune diseases mediated by pathogenic IgG autoantibodies. Engineered for increased FcRn affinity at both acidic and physiological pH, efgartigimod can outcompete endogenous IgG binding, preventing FcRn-mediated recycling of IgGs and resulting in increased lysosomal degradation. A population pharmacokinetic-pharmacodynamic (PKPD) model including FcRn binding was developed based on data from two healthy volunteer studies after single and repeated administration of efgartigimod. This model was able to simultaneously describe the serum efgartigimod and total IgG profiles across dose groups, using drug-induced FcRn receptor occupancy as driver of total IgG suppression. The model was expanded to describe the PKPD of efgartigimod in cynomolgus monkeys, rabbits, rats and mice. Most species differences were explainable by including the species-specific in vitro affinity for FcRn binding at pH 7.4 and by allometric scaling of the physiological parameters. In vitro-in vivo scaling proved crucial for translation success: the drug effect was over/underpredicted in rabbits/mice when ignoring the lower/higher binding affinity of efgartigimod for these species versus human, respectively. Given the successful model prediction of the PK and total IgG dynamics across species, it was concluded that the PKPD of efgartigimod can be characterized by target binding. From the model, it is suggested that the initial fast decrease of measurable unbound efgartigimod following dosing is the result of combined clearance of free drug and high affinity target binding, while the relatively slow terminal PK phase reflects release of bound drug from the receptor. High affinity target binding protects the drug from elimination and results in a sustained PD effect characterized by an increase in the IgG degradation rate constant with increasing target receptor occupancy.</p>","PeriodicalId":16851,"journal":{"name":"Journal of Pharmacokinetics and Pharmacodynamics","volume":"52 1","pages":"2"},"PeriodicalIF":2.2,"publicationDate":"2024-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11621151/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142785792","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Semi-mechanistic population pharmacokinetic modeling of DZIF-10c, a neutralizing antibody against SARS-Cov-2: predicting systemic and lung exposure following inhaled and intravenous administration.
IF 2.2 4区 医学
Journal of Pharmacokinetics and Pharmacodynamics Pub Date : 2024-12-05 DOI: 10.1007/s10928-024-09947-2
Sree Kurup, Nieves Velez de Mendizabal, Stephan Becker, Erica Bolella, Dorothy De Sousa, Gerd Fätkenheuer, Henning Gruell, Florian Klein, Jakob J Malin, Ulrike Schmid, Julia Korell
{"title":"Semi-mechanistic population pharmacokinetic modeling of DZIF-10c, a neutralizing antibody against SARS-Cov-2: predicting systemic and lung exposure following inhaled and intravenous administration.","authors":"Sree Kurup, Nieves Velez de Mendizabal, Stephan Becker, Erica Bolella, Dorothy De Sousa, Gerd Fätkenheuer, Henning Gruell, Florian Klein, Jakob J Malin, Ulrike Schmid, Julia Korell","doi":"10.1007/s10928-024-09947-2","DOIUrl":"10.1007/s10928-024-09947-2","url":null,"abstract":"<p><p>DZIF-10c (BI 767551) is a recombinant human monoclonal antibody of the IgG1 kappa isotype. It acts as a SARS-CoV-2 neutralizing antibody. DZIF-10c has been developed for both systemic exposure by intravenous infusion as well as for specific exposure to the respiratory tract by application as an inhaled aerosol generated by a nebulizer. An integrated preclinical/clinical semi-mechanistic population pharmacokinetic model was developed to characterize the exposure profile of DZIF-10c in the systemic circulation and lungs. To inform and reduce uncertainty around exposure in the lungs following different methods of dosing, preclinical cynomolgus monkey data was combined with human data using allometric scaling principles. Human serum concentrations of DZIF-10c from two clinical trials were combined with serum/plasma and lung epithelial lining fluid (ELF) concentrations from three preclinical studies to characterize the relationship between dosing, serum/plasma, and lung exposure. The final model was used to predict exposure in the lungs following different routes of administration. Simulations showed that inhalation provides immediate and relevant exposure in the lung ELF at a much lower dose compared with an infusion. Combining inhalation with intravenous therapy results in high and sustained DZIF-10c exposure in the lungs and systemic circulation, thereby combining the benefits of both routes of administration. By combining preclinical data with clinical data (via allometric scaling principles), the developed population pharmacokinetic model reduced uncertainty around exposure in the lungs allowing evaluation of alternative dosing strategies to achieve the desired concentrations of DZIF-10c in human lungs.</p>","PeriodicalId":16851,"journal":{"name":"Journal of Pharmacokinetics and Pharmacodynamics","volume":"52 1","pages":"3"},"PeriodicalIF":2.2,"publicationDate":"2024-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11621205/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142785785","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Multiorgan-on-a-chip for cancer drug pharmacokinetics-pharmacodynamics (PK-PD) modeling and simulations.
IF 2.2 4区 医学
Journal of Pharmacokinetics and Pharmacodynamics Pub Date : 2024-12-04 DOI: 10.1007/s10928-024-09955-2
Abdurehman Eshete Mohammed, Filiz Kurucaovalı, Devrim Pesen Okvur
{"title":"Multiorgan-on-a-chip for cancer drug pharmacokinetics-pharmacodynamics (PK-PD) modeling and simulations.","authors":"Abdurehman Eshete Mohammed, Filiz Kurucaovalı, Devrim Pesen Okvur","doi":"10.1007/s10928-024-09955-2","DOIUrl":"https://doi.org/10.1007/s10928-024-09955-2","url":null,"abstract":"<p><p>Cancer is one of the most common and fatal diseases worldwide and kills millions of people every year. Cancer drug resistance, lack of efficacy, and safety are significant problems in cancer patients. A multiorgan-on-a-chip (MOC) device consisting of breast and liver compartments was designed with AutoCAD software. The MOC molds were printed by a Formlabs Form 2 3D printer. MDA-MB-231, HepG2, and MCF-10 A cells were used for the MOC experiments. The cell lines were cultured at 37 °C with 5% CO<sub>2,</sub> and cell viability was assessed via Alamar blue dye to generate pharmacodynamics (PD) data. Drug concentrations from the cell culture media were analyzed via Agilent 1260 Infinity II HPLC with a Waters Symmetry C18 column and used to generate pharmacokinetics (PK) data. The PK and PD data were modeled and simulated by Monolix and Simulix software, respectively. The safety and efficacy of drug dosing regimens were compared, and the best dosing regimens were selected. This research designed and fabricated a unique MOC consisting of liver and breast compartments that overcomes the need for sealing or assembling. It was used for PK-PD modeling and simulations, and its functionality was proven experimentally. The new MOC will be helpful in preclinical trials to evaluate the efficacy and safety of drugs.</p>","PeriodicalId":16851,"journal":{"name":"Journal of Pharmacokinetics and Pharmacodynamics","volume":"52 1","pages":"1"},"PeriodicalIF":2.2,"publicationDate":"2024-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142770065","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mixed effect estimation in deep compartment models: Variational methods outperform first-order approximations. 深隔室模型中的混合效应估计:变量方法优于一阶近似方法
IF 2.2 4区 医学
Journal of Pharmacokinetics and Pharmacodynamics Pub Date : 2024-12-01 Epub Date: 2024-07-04 DOI: 10.1007/s10928-024-09931-w
Alexander Janssen, Frank C Bennis, Marjon H Cnossen, Ron A A Mathôt
{"title":"Mixed effect estimation in deep compartment models: Variational methods outperform first-order approximations.","authors":"Alexander Janssen, Frank C Bennis, Marjon H Cnossen, Ron A A Mathôt","doi":"10.1007/s10928-024-09931-w","DOIUrl":"10.1007/s10928-024-09931-w","url":null,"abstract":"<p><p>This work focusses on extending the deep compartment model (DCM) framework to the estimation of mixed-effects. By introducing random effects, model predictions can be personalized based on drug measurements, enabling the testing of different treatment schedules on an individual basis. The performance of classical first-order (FO and FOCE) and machine learning based variational inference (VI) algorithms were compared in a simulation study. In VI, posterior distributions of the random variables are approximated using variational distributions whose parameters can be directly optimized. We found that variational approximations estimated using the path derivative gradient estimator version of VI were highly accurate. Models fit on the simulated data set using the FO and VI objective functions gave similar results, with accurate predictions of both the population parameters and covariate effects. Contrastingly, models fit using FOCE depicted erratic behaviour during optimization, and resulting parameter estimates were inaccurate. Finally, we compared the performance of the methods on two real-world data sets of haemophilia A patients who received standard half-life factor VIII concentrates during prophylactic and perioperative settings. Again, models fit using FO and VI depicted similar results, although some models fit using FO presented divergent results. Again, models fit using FOCE were unstable. In conclusion, we show that mixed-effects estimation using the DCM is feasible. VI performs conditional estimation, which might lead to more accurate results in more complex models compared to the FO method.</p>","PeriodicalId":16851,"journal":{"name":"Journal of Pharmacokinetics and Pharmacodynamics","volume":" ","pages":"797-808"},"PeriodicalIF":2.2,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11579087/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141534587","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
0
×
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×
相关产品
×
本文献相关产品
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