首页 > 最新文献

Drug Metabolism and Disposition最新文献

英文 中文
Corrigendum to "Pharmacokinetic/Pharmacodynamic Assessment of the Structural Refinement of Clopidogrel Focusing on the Balance between Bioactivation and Deactivation".
IF 4.4 3区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-04-04 DOI: 10.1016/j.dmd.2025.100079
{"title":"Corrigendum to \"Pharmacokinetic/Pharmacodynamic Assessment of the Structural Refinement of Clopidogrel Focusing on the Balance between Bioactivation and Deactivation\".","authors":"","doi":"10.1016/j.dmd.2025.100079","DOIUrl":"https://doi.org/10.1016/j.dmd.2025.100079","url":null,"abstract":"","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 4","pages":"100079"},"PeriodicalIF":4.4,"publicationDate":"2025-04-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143788128","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}
引用次数: 0
Human absorption, distribution, metabolism, and excretion studies: Conventional or microtracer?
IF 4.4 3区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-03-19 DOI: 10.1016/j.dmd.2025.100067
Sean Xiaochun Zhu

A human absorption, distribution, metabolism, and excretion (hADME) study is an essential clinical pharmacology study for small-molecule drugs. The study provides insights into circulating drug-related materials and the drug's elimination pathways in humans, which can guide future studies on safety and drug-drug interaction of metabolites as well as organ impairment and drug-drug interaction of the parent drug. The 2 hADME study types, namely conventional and microtracer, are comprehensively compared in this manuscript. A review of literature found that conventional hADME studies were approximately 7 times that of microtracer hADME studies for small molecule and peptide drugs based on publications in 3 peer-reviewed journals from 2010 to 2024. Each study type has advantages and disadvantages. The advantages of conventional hADME studies primarily include the ease, low cost, and flexibility of radiometric sample analysis. In contrast, the advantages of microtracer hADME studies primarily include exemption from prerequisite studies and use of non-good manufacturing practice 14C-labeled materials. The disadvantages of each study type are essentially the advantages of the other. The manuscript also discusses scenarios where a microtracer hADME study may be preferable. Finally, recommendations are provided on selecting the appropriate hADME study type for an investigational drug. SIGNIFICANCE STATEMENT: The manuscript discusses 2 primary human absorption, distribution, metabolism, and excretion study types: conventional and microtracer. It covers published literature studies, the pros and cons of each type, scenarios for conducting microtracer studies, and a recommended decision tree for selecting the appropriate human absorption, distribution, metabolism, and excretion study type.

人体吸收、分布、代谢和排泄(hADME)研究是小分子药物必不可少的临床药理研究。通过这项研究,可以深入了解药物在人体内的循环相关物质和药物的消除途径,从而为今后研究代谢物的安全性和药物相互作用以及母体药物的器官损害和药物相互作用提供指导。本手稿全面比较了两种 hADME 研究类型,即传统型和微示踪型。文献综述发现,根据2010年至2024年在3本同行评审期刊上发表的论文,在小分子和多肽药物方面,常规hADME研究约为微示踪hADME研究的7倍。每种研究类型都各有利弊。传统 hADME 研究的优点主要包括辐射样品分析的简便性、低成本和灵活性。相比之下,微量示踪法氢残留检测研究的优点主要包括无需进行先决研究和使用非良好生产规范的 14C 标记材料。每种研究类型的缺点基本上都是对方的优点。手稿还讨论了在哪些情况下微量示踪剂 hADME 研究可能更可取。最后,还就如何为研究药物选择合适的 hADME 研究类型提出了建议。意义声明:本手稿讨论了两种主要的人体吸收、分布、代谢和排泄研究类型:传统型和微量示踪剂型。它涵盖了已发表的文献研究、每种类型的优缺点、进行微示踪剂研究的情景,以及选择适当的人体吸收、分布、代谢和排泄研究类型的建议决策树。
{"title":"Human absorption, distribution, metabolism, and excretion studies: Conventional or microtracer?","authors":"Sean Xiaochun Zhu","doi":"10.1016/j.dmd.2025.100067","DOIUrl":"https://doi.org/10.1016/j.dmd.2025.100067","url":null,"abstract":"<p><p>A human absorption, distribution, metabolism, and excretion (hADME) study is an essential clinical pharmacology study for small-molecule drugs. The study provides insights into circulating drug-related materials and the drug's elimination pathways in humans, which can guide future studies on safety and drug-drug interaction of metabolites as well as organ impairment and drug-drug interaction of the parent drug. The 2 hADME study types, namely conventional and microtracer, are comprehensively compared in this manuscript. A review of literature found that conventional hADME studies were approximately 7 times that of microtracer hADME studies for small molecule and peptide drugs based on publications in 3 peer-reviewed journals from 2010 to 2024. Each study type has advantages and disadvantages. The advantages of conventional hADME studies primarily include the ease, low cost, and flexibility of radiometric sample analysis. In contrast, the advantages of microtracer hADME studies primarily include exemption from prerequisite studies and use of non-good manufacturing practice <sup>14</sup>C-labeled materials. The disadvantages of each study type are essentially the advantages of the other. The manuscript also discusses scenarios where a microtracer hADME study may be preferable. Finally, recommendations are provided on selecting the appropriate hADME study type for an investigational drug. SIGNIFICANCE STATEMENT: The manuscript discusses 2 primary human absorption, distribution, metabolism, and excretion study types: conventional and microtracer. It covers published literature studies, the pros and cons of each type, scenarios for conducting microtracer studies, and a recommended decision tree for selecting the appropriate human absorption, distribution, metabolism, and excretion study type.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 5","pages":"100067"},"PeriodicalIF":4.4,"publicationDate":"2025-03-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143810735","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}
引用次数: 0
In vitro-in vivo scaling of cytochrome P450-mediated metabolic clearance using a relative activity factor approach.
IF 4.4 3区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-03-14 DOI: 10.1016/j.dmd.2025.100065
Yoko Nagaya, Yoshitane Nozaki

Quantitative prediction of hepatic clearance is a key element in predicting the human pharmacokinetic profile in the nonclinical stages. In the present study, we focused on the major cytochrome P450 (P450) isoforms (CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4) and tested a relative activity factor (RAF) method to quantitatively predict in vivo hepatic intrinsic clearance (CLh,int,vivo) and fraction metabolized (fm) by the P450 isoforms directly from an in vitro recombinant P450 system. We selected multiple probe substrates for CYP1A2 (caffeine, tizanidine, phenacetin), CYP2C9 ((S)-acenocoumarol, glimepiride, lornoxicam, tolbutamide, (S)-warfarin), CYP2C19 ((S)-lansoprazole, omeprazole, pantoprazole), CYP2D6 (desipramine, metoprolol, nebivolol, tolterodine), and CYP3A4 (alprazolam, felodipine, midazolam, nisoldipine, sildenafil, triazolam) to calculate the representative RAF value for each P450 isoform based on the in vivo-to-in vitro clearance ratio of the multiple probe substrates. The most pronounced substrate dependency of the RAF values was noted for CYP3A4 (2698 [alprazolam] to 19073 [nisoldipine] pmol P450/kg). Using the geometric mean of the RAF values for each isoform, a within 3-fold prediction of the CLh,int,vivo was obtained for all the 11 test drugs, except glibenclamide, which is a known substrate of hepatic uptake transporters. The fm values of the responsible P450 isoform(s) could be well predicted for mexiletine, tamsulosin, risperidone, celecoxib, and glibenclamide. This simple, practical RAF method can be one of the useful nonclinical methods to estimate the CLh,int,vivo and fm mediated by the major P450 isoforms, which would promote earlier understanding of the impact of genetic polymorphisms and drug-drug interactions on the human pharmacokinetics of the substrate compounds. SIGNIFICANCE STATEMENT: The relative activity factor method has been used for extrapolating in vitro clearance from recombinant systems to liver microsomes, but this study utilized this method to predict in vivo hepatic clearance and fraction metabolized values. By applying relative activity factor values obtained from multiple probe substrates, this study was able to quantitatively predict the in vivo clearances mediated by CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4. This simple, practical method will help optimize metabolic clearances via the major cytochrome P450 isoforms in the nonclinical stages.

肝清除率的定量预测是非临床阶段预测人体药代动力学特征的关键因素。在本研究中,我们重点研究了主要的细胞色素 P450(P450)同工酶(CYP1A2、CYP2C9、CYP2C19、CYP2D6、CYP3A4),并测试了一种相对活性因子(RAF)方法,以直接从体外重组 P450 系统中定量预测体内肝脏固有清除率(CLh,int,vivo)和 P450 同工酶的代谢分数(fm)。我们选择了 CYP1A2(咖啡因、替扎尼丁、苯乙酸)、CYP2C9((S)-醋硝香豆素、格列美脲、洛诺昔康、托布他胺、(S)-华法林)、CYP2C19((S)-兰索拉唑、奥美拉唑、泮托拉唑)、CYP2D6(地西帕明、美托洛尔、奈博洛尔、托布他胺、(S)-华法林)的多种探针底物、CYP2D6(地西帕明、美托洛尔、托特罗定)和 CYP3A4(阿普唑仑、非洛地平、咪达唑仑、尼索地平、西地那非、三唑仑),根据多种探针底物的体内与体外清除率比值计算出每种 P450 同工酶的代表性 RAF 值。CYP3A4 的 RAF 值与底物的关系最为明显(2698 [阿普唑仑] 至 19073 [尼索地平] pmol P450/kg)。利用每种同工酶的 RAF 值的几何平均数,可以预测所有 11 种测试药物的体内 CLh 值在 3 倍以内,但格列本脲(已知是肝脏摄取转运体的底物)除外。对于美西列汀、坦索罗辛、利培酮、塞来昔布和格列本脲,可以很好地预测负责的 P450 同工酶的 fm 值。这种简单实用的 RAF 方法可以作为估算主要 P450 同工酶介导的 CLh、int、vivo 和 fm 的有用非临床方法之一,从而有助于更早地了解基因多态性和药物间相互作用对底物化合物人体药代动力学的影响。意义说明:相对活性因子法一直用于将重组系统的体外清除率外推至肝脏微粒体,但本研究利用该方法预测了体内肝脏清除率和代谢分数值。通过应用从多个探针底物中获得的相对活性因子值,本研究能够定量预测由 CYP1A2、CYP2C9、CYP2C19、CYP2D6 和 CYP3A4 介导的体内清除率。这种简单实用的方法有助于优化非临床阶段通过主要细胞色素 P450 同工酶的代谢清除率。
{"title":"In vitro-in vivo scaling of cytochrome P450-mediated metabolic clearance using a relative activity factor approach.","authors":"Yoko Nagaya, Yoshitane Nozaki","doi":"10.1016/j.dmd.2025.100065","DOIUrl":"https://doi.org/10.1016/j.dmd.2025.100065","url":null,"abstract":"<p><p>Quantitative prediction of hepatic clearance is a key element in predicting the human pharmacokinetic profile in the nonclinical stages. In the present study, we focused on the major cytochrome P450 (P450) isoforms (CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP3A4) and tested a relative activity factor (RAF) method to quantitatively predict in vivo hepatic intrinsic clearance (CL<sub>h,int,vivo</sub>) and fraction metabolized (f<sub>m</sub>) by the P450 isoforms directly from an in vitro recombinant P450 system. We selected multiple probe substrates for CYP1A2 (caffeine, tizanidine, phenacetin), CYP2C9 ((S)-acenocoumarol, glimepiride, lornoxicam, tolbutamide, (S)-warfarin), CYP2C19 ((S)-lansoprazole, omeprazole, pantoprazole), CYP2D6 (desipramine, metoprolol, nebivolol, tolterodine), and CYP3A4 (alprazolam, felodipine, midazolam, nisoldipine, sildenafil, triazolam) to calculate the representative RAF value for each P450 isoform based on the in vivo-to-in vitro clearance ratio of the multiple probe substrates. The most pronounced substrate dependency of the RAF values was noted for CYP3A4 (2698 [alprazolam] to 19073 [nisoldipine] pmol P450/kg). Using the geometric mean of the RAF values for each isoform, a within 3-fold prediction of the CL<sub>h,int,vivo</sub> was obtained for all the 11 test drugs, except glibenclamide, which is a known substrate of hepatic uptake transporters. The f<sub>m</sub> values of the responsible P450 isoform(s) could be well predicted for mexiletine, tamsulosin, risperidone, celecoxib, and glibenclamide. This simple, practical RAF method can be one of the useful nonclinical methods to estimate the CL<sub>h,int,vivo</sub> and f<sub>m</sub> mediated by the major P450 isoforms, which would promote earlier understanding of the impact of genetic polymorphisms and drug-drug interactions on the human pharmacokinetics of the substrate compounds. SIGNIFICANCE STATEMENT: The relative activity factor method has been used for extrapolating in vitro clearance from recombinant systems to liver microsomes, but this study utilized this method to predict in vivo hepatic clearance and fraction metabolized values. By applying relative activity factor values obtained from multiple probe substrates, this study was able to quantitatively predict the in vivo clearances mediated by CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4. This simple, practical method will help optimize metabolic clearances via the major cytochrome P450 isoforms in the nonclinical stages.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 4","pages":"100065"},"PeriodicalIF":4.4,"publicationDate":"2025-03-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143810733","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}
引用次数: 0
CYP1A2 contributes to the metabolism of mefloquine: Exploration using in vitro metabolism and physiologically-based pharmacokinetic modelling.
IF 4.4 3区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-03-08 DOI: 10.1016/j.dmd.2025.100060
Cleavon K Cloete, Preshendren Govender, Nicholas Njuguna, Neil J Parrott, Kenichi Umehara, Kelly Chibale, Mathew Njoroge

Mefloquine is an antimalarial drug routinely used for prophylaxis and in the treatment of malaria. Approximately 50% of mefloquine metabolism, both in vitro and in vivo, is mediated by CYP3A4 with the remaining contributions by other CYP450 isoforms unaccounted for. This study aimed to determine the contribution of CYP450s to mefloquine metabolism and incorporate this knowledge into a physiologically-based pharmacokinetic model. The data in human liver microsomes demonstrated the involvement of CYP3A4/5 as well as the previously unreported contribution of CYP1A2 to mefloquine metabolism. The fraction metabolized by CYP1A2 (fm,CYP1A2) was estimated to be at least 50% using chemical inhibitors and pooled human liver microsomes and confirmed using recombinant human CYP450 enzymes. A physiologically-based pharmacokinetic model built in Simcyp using the fm,CYP values recaptured observed clinical pharmacokinetic data-71 % of the simulated area under the curve (AUC) values were within 1.25-fold of the observed clinical data, and all simulated AUC values were within 2-fold of observed data. Simulated mefloquine exposures increased by 88% when an interaction with fluvoxamine, a CYP1A2 inhibitor was modeled. Modeling showed that heavy smoking, and subsequent induction of CYP1A2, had a notable effect on mefloquine exposure. CYP1A2 genotype status also influenced mefloquine exposure with a predicted AUC ratio of 1.68 in a simulated population of CYP1A2 poor metabolizers. The involvement of CYP1A2 in mefloquine metabolism suggests a previously unreported drug-drug interaction risk. Looking forward, the analysis here suggests the clinical exploration of the interaction between mefloquine and CYP1A2. SIGNIFICANCE STATEMENT: Despite the widespread use of mefloquine in malaria prophylaxis and treatment, its metabolism is not completely characterized. This has implications for understanding and predicting drug-drug interactions involving mefloquine. Here, we identify CYP1A2 as a key enzyme involved in mefloquine metabolism and use physiologically-based pharmacokinetic modeling to demonstrate the contribution of this route to interactions with mefloquine. The in vitro data and revised physiologically-based pharmacokinetic model are important starting points for future exploration of this pathway using clinical data.

{"title":"CYP1A2 contributes to the metabolism of mefloquine: Exploration using in vitro metabolism and physiologically-based pharmacokinetic modelling.","authors":"Cleavon K Cloete, Preshendren Govender, Nicholas Njuguna, Neil J Parrott, Kenichi Umehara, Kelly Chibale, Mathew Njoroge","doi":"10.1016/j.dmd.2025.100060","DOIUrl":"https://doi.org/10.1016/j.dmd.2025.100060","url":null,"abstract":"<p><p>Mefloquine is an antimalarial drug routinely used for prophylaxis and in the treatment of malaria. Approximately 50% of mefloquine metabolism, both in vitro and in vivo, is mediated by CYP3A4 with the remaining contributions by other CYP450 isoforms unaccounted for. This study aimed to determine the contribution of CYP450s to mefloquine metabolism and incorporate this knowledge into a physiologically-based pharmacokinetic model. The data in human liver microsomes demonstrated the involvement of CYP3A4/5 as well as the previously unreported contribution of CYP1A2 to mefloquine metabolism. The fraction metabolized by CYP1A2 (fm,CYP1A2) was estimated to be at least 50% using chemical inhibitors and pooled human liver microsomes and confirmed using recombinant human CYP450 enzymes. A physiologically-based pharmacokinetic model built in Simcyp using the fm,CYP values recaptured observed clinical pharmacokinetic data-71 % of the simulated area under the curve (AUC) values were within 1.25-fold of the observed clinical data, and all simulated AUC values were within 2-fold of observed data. Simulated mefloquine exposures increased by 88% when an interaction with fluvoxamine, a CYP1A2 inhibitor was modeled. Modeling showed that heavy smoking, and subsequent induction of CYP1A2, had a notable effect on mefloquine exposure. CYP1A2 genotype status also influenced mefloquine exposure with a predicted AUC ratio of 1.68 in a simulated population of CYP1A2 poor metabolizers. The involvement of CYP1A2 in mefloquine metabolism suggests a previously unreported drug-drug interaction risk. Looking forward, the analysis here suggests the clinical exploration of the interaction between mefloquine and CYP1A2. SIGNIFICANCE STATEMENT: Despite the widespread use of mefloquine in malaria prophylaxis and treatment, its metabolism is not completely characterized. This has implications for understanding and predicting drug-drug interactions involving mefloquine. Here, we identify CYP1A2 as a key enzyme involved in mefloquine metabolism and use physiologically-based pharmacokinetic modeling to demonstrate the contribution of this route to interactions with mefloquine. The in vitro data and revised physiologically-based pharmacokinetic model are important starting points for future exploration of this pathway using clinical data.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 4","pages":"100060"},"PeriodicalIF":4.4,"publicationDate":"2025-03-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143788130","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}
引用次数: 0
Novel O-methylpyrimidine prodrugs of phenolic compounds bioactivated by aldehyde oxidase: Enhancing metabolic stability against first-pass conjugative metabolism in the intestine.
IF 4.4 3区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-03-05 DOI: 10.1016/j.dmd.2025.100059
John P Kowalski, Brian R Baer, Samuel D Randall, Karin Brown, Amy Crooks, Joseph McCown, Matthew G McDonald, Jackie Harrison, Suomia Abuirqeba, Donghua Dai, Michael Hilton, James T Brewster, Alex A Kellum

Phenol-containing drugs may exhibit limited oral bioavailability due to first-pass conjugation in the intestine and liver, and potentially unfavorable biopharmaceutical properties imparted by the hydrogen-bond donor. We present a novel prodrug strategy in which O-methylpyrimidine modification masks the phenolic moiety and employs aldehyde oxidase (AO) to release the parent drug. Prototypical prodrugs of 4-hydroxy-tamoxifen (4OH-TAM), raloxifene (RAL), rotigotine, 5-hydroxy-tolterodine, and phentolamine were all substrates for AO-mediated parent drug release in liver cytosol from humans and every preclinical species evaluated. Reaction phenotyping confirmed the role of AO; hydralazine inhibited production of 4OH-TAM and RAL from their respective prodrugs in the human liver cytosol, and recombinant human AO activated those same prodrugs. Based on the identified byproduct, 5-(hydroxymethyl)uracil, and characterized 4OH-TAM prodrug metabolite intermediates, a mechanism is proposed, involving oxidation of the pyrimidine 4-position, followed by rate-limiting oxidation at the 2-position and subsequent C-O bond cleavage via an imine-methide intermediate. To determine a preclinical animal for proof-of-concept prodrug activation in vivo, we measured both absolute AO protein concentration and parent release for 2 prodrugs in the liver cytosol of multiple species and found that hamster was a promising candidate to model humans. After confirming a similar balance of AO-mediated prodrug conversion versus nonproductive/subsequent biotransformation in human and hamster hepatocytes, the 4OH-TAM prodrug and RAL prodrug 1 were progressed to a pharmacokinetic study in hamsters. A 30 mg/kg oral dose of RAL prodrug 1 demonstrated a 2-fold increase in RAL exposure compared with dosing parent RAL, indicating that this novel prodrug strategy has the potential to improve bioavailability in humans. SIGNIFICANCE STATEMENT: An aldehyde oxidase-mediated biotransformation that cleaves O-linked methylpyrimidine-masked phenolic moieties was identified, and this system employed for a novel prodrug bioactivation strategy. The research herein expands existing knowledge surrounding the metabolism capabilities of this enzyme and provides medicinal chemists with a tool to enhance the oral bioavailability of phenolic compounds that otherwise would be limited due to extensive phase II metabolism and possibly low permeability.

{"title":"Novel O-methylpyrimidine prodrugs of phenolic compounds bioactivated by aldehyde oxidase: Enhancing metabolic stability against first-pass conjugative metabolism in the intestine.","authors":"John P Kowalski, Brian R Baer, Samuel D Randall, Karin Brown, Amy Crooks, Joseph McCown, Matthew G McDonald, Jackie Harrison, Suomia Abuirqeba, Donghua Dai, Michael Hilton, James T Brewster, Alex A Kellum","doi":"10.1016/j.dmd.2025.100059","DOIUrl":"https://doi.org/10.1016/j.dmd.2025.100059","url":null,"abstract":"<p><p>Phenol-containing drugs may exhibit limited oral bioavailability due to first-pass conjugation in the intestine and liver, and potentially unfavorable biopharmaceutical properties imparted by the hydrogen-bond donor. We present a novel prodrug strategy in which O-methylpyrimidine modification masks the phenolic moiety and employs aldehyde oxidase (AO) to release the parent drug. Prototypical prodrugs of 4-hydroxy-tamoxifen (4OH-TAM), raloxifene (RAL), rotigotine, 5-hydroxy-tolterodine, and phentolamine were all substrates for AO-mediated parent drug release in liver cytosol from humans and every preclinical species evaluated. Reaction phenotyping confirmed the role of AO; hydralazine inhibited production of 4OH-TAM and RAL from their respective prodrugs in the human liver cytosol, and recombinant human AO activated those same prodrugs. Based on the identified byproduct, 5-(hydroxymethyl)uracil, and characterized 4OH-TAM prodrug metabolite intermediates, a mechanism is proposed, involving oxidation of the pyrimidine 4-position, followed by rate-limiting oxidation at the 2-position and subsequent C-O bond cleavage via an imine-methide intermediate. To determine a preclinical animal for proof-of-concept prodrug activation in vivo, we measured both absolute AO protein concentration and parent release for 2 prodrugs in the liver cytosol of multiple species and found that hamster was a promising candidate to model humans. After confirming a similar balance of AO-mediated prodrug conversion versus nonproductive/subsequent biotransformation in human and hamster hepatocytes, the 4OH-TAM prodrug and RAL prodrug 1 were progressed to a pharmacokinetic study in hamsters. A 30 mg/kg oral dose of RAL prodrug 1 demonstrated a 2-fold increase in RAL exposure compared with dosing parent RAL, indicating that this novel prodrug strategy has the potential to improve bioavailability in humans. SIGNIFICANCE STATEMENT: An aldehyde oxidase-mediated biotransformation that cleaves O-linked methylpyrimidine-masked phenolic moieties was identified, and this system employed for a novel prodrug bioactivation strategy. The research herein expands existing knowledge surrounding the metabolism capabilities of this enzyme and provides medicinal chemists with a tool to enhance the oral bioavailability of phenolic compounds that otherwise would be limited due to extensive phase II metabolism and possibly low permeability.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 4","pages":"100059"},"PeriodicalIF":4.4,"publicationDate":"2025-03-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143788131","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}
引用次数: 0
Switch/sucrose non-fermentable complex interacts with constitutive androstane receptor to regulate drug-metabolizing enzymes and transporters in the liver.
IF 4.4 3区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-03-04 DOI: 10.1016/j.dmd.2025.100057
Kiamu Kurosawa, Masataka Nakano, Itsuki Yokoseki, Mei Tomii, Yuichiro Higuchi, Shotaro Uehara, Nao Yoneda, Hiroshi Suemizu, Tatsuki Fukami, Miki Nakajima

Constitutive androstane receptor (CAR) is a nuclear receptor that plays an important role in regulating drug metabolism and bile acid homeostasis in the liver. Recently, it was revealed that the switch/sucrose non-fermentable (SWI/SNF) complex, a chromatin remodeler, regulates transactivation by nuclear receptors, such as the pregnane X receptor and vitamin D receptor. However, studies on the involvement of the SWI/SNF complex in CAR-mediated transactivation are limited. Here, we demonstrated that the induction of cytochrome P450 CYP2B6 expression by CAR activators, 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime and phenobarbital, was enhanced by the inhibition of AT-rich interactive domain-containing protein (ARID) 1A, a canonical brahma-related gene 1-associated factor (cBAF) component, one of the SWI/SNF complexes, and was attenuated by inhibition of bromodomain-containing protein (BRD) 9, a noncanonical BAF (ncBAF) component, in primary hepatocytes from humanized mice. Coimmunoprecipitation assays revealed that ARID1A and BRD9 interacted with CAR. Chromatin immunoprecipitation assay revealed that the 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime-induced binding of CAR to the 5'-flanking region of CYP2B6 gene increased with ARID1A inhibition and reduced with BRD9 inhibition. These results suggest that cBAF negatively regulates CAR-mediated transactivation by attenuating CAR binding to its response element, whereas ncBAF positively regulates it by facilitating CAR binding. Furthermore, ARID1A inhibition enhanced phenobarbital-induced increases in UDP-glucuronosyltransferase 1A1 expression and multidrug resistance-associated protein 2 mRNA level and activity. Collectively, our findings indicate that cBAF and ncBAF play essential roles in xenobiotic metabolism by regulating CAR-mediated transactivation and that ARID1A inhibitors may offer therapeutic benefits for hyperbilirubinemia and cholestasis by inducing UDP-glucuronosyltransferase 1A1 and multidrug resistance-associated protein 2 expression. SIGNIFICANCE STATEMENT: This study revealed that canonical brahma-related gene 1-associated factor and noncanonical brahma-related gene 1-associated factor, members of the switch/sucrose non-fermentable family, negatively and positively regulate constitutive androstane receptor (CAR) transactivation, respectively, through changes in the chromatin structure around the CAR response element in the 5'-flanking regions of CAR target genes. The inhibition of AT-rich interactive domain-containing protein 1A may be beneficial for cholestasis treatment by enhancing CAR-mediated transactivation.

{"title":"Switch/sucrose non-fermentable complex interacts with constitutive androstane receptor to regulate drug-metabolizing enzymes and transporters in the liver.","authors":"Kiamu Kurosawa, Masataka Nakano, Itsuki Yokoseki, Mei Tomii, Yuichiro Higuchi, Shotaro Uehara, Nao Yoneda, Hiroshi Suemizu, Tatsuki Fukami, Miki Nakajima","doi":"10.1016/j.dmd.2025.100057","DOIUrl":"https://doi.org/10.1016/j.dmd.2025.100057","url":null,"abstract":"<p><p>Constitutive androstane receptor (CAR) is a nuclear receptor that plays an important role in regulating drug metabolism and bile acid homeostasis in the liver. Recently, it was revealed that the switch/sucrose non-fermentable (SWI/SNF) complex, a chromatin remodeler, regulates transactivation by nuclear receptors, such as the pregnane X receptor and vitamin D receptor. However, studies on the involvement of the SWI/SNF complex in CAR-mediated transactivation are limited. Here, we demonstrated that the induction of cytochrome P450 CYP2B6 expression by CAR activators, 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime and phenobarbital, was enhanced by the inhibition of AT-rich interactive domain-containing protein (ARID) 1A, a canonical brahma-related gene 1-associated factor (cBAF) component, one of the SWI/SNF complexes, and was attenuated by inhibition of bromodomain-containing protein (BRD) 9, a noncanonical BAF (ncBAF) component, in primary hepatocytes from humanized mice. Coimmunoprecipitation assays revealed that ARID1A and BRD9 interacted with CAR. Chromatin immunoprecipitation assay revealed that the 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime-induced binding of CAR to the 5'-flanking region of CYP2B6 gene increased with ARID1A inhibition and reduced with BRD9 inhibition. These results suggest that cBAF negatively regulates CAR-mediated transactivation by attenuating CAR binding to its response element, whereas ncBAF positively regulates it by facilitating CAR binding. Furthermore, ARID1A inhibition enhanced phenobarbital-induced increases in UDP-glucuronosyltransferase 1A1 expression and multidrug resistance-associated protein 2 mRNA level and activity. Collectively, our findings indicate that cBAF and ncBAF play essential roles in xenobiotic metabolism by regulating CAR-mediated transactivation and that ARID1A inhibitors may offer therapeutic benefits for hyperbilirubinemia and cholestasis by inducing UDP-glucuronosyltransferase 1A1 and multidrug resistance-associated protein 2 expression. SIGNIFICANCE STATEMENT: This study revealed that canonical brahma-related gene 1-associated factor and noncanonical brahma-related gene 1-associated factor, members of the switch/sucrose non-fermentable family, negatively and positively regulate constitutive androstane receptor (CAR) transactivation, respectively, through changes in the chromatin structure around the CAR response element in the 5'-flanking regions of CAR target genes. The inhibition of AT-rich interactive domain-containing protein 1A may be beneficial for cholestasis treatment by enhancing CAR-mediated transactivation.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 4","pages":"100057"},"PeriodicalIF":4.4,"publicationDate":"2025-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143751325","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}
引用次数: 0
Development of human embryonic stem cell-derived cardiomyocytes and application of fluorescence probe substrate for characterization of cytochrome P450 enzyme 2J2.
IF 4.4 3区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-03-03 DOI: 10.1016/j.dmd.2025.100053
Xingyu Zhu, Yee Kiat Soh, Mingxin Wan, Jeremy Kah Sheng Pang, Wei Liang Leow, Chong Tian, Boon Seng Soh, Eric Chun Yong Chan

Cardiac cytochrome P450 2J2 (CYP2J2) plays a significant role in cardiovascular homeostasis due to its dual functions in drug metabolism and the epoxidation of polyunsaturated fatty acids. Additionally, the inhibition of CYP2J2 by xenobiotics has been linked to drug-induced cardiotoxicity, warranting further investigation of this critical enzyme in cardiac systems. Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) are physiologically relevant in vitro models that recapitulate relevant phenotypes important for cardiovascular research. However, no studies have so far characterized CYP2J2 expression and activities in these models. Here, we developed and validated H7 hESC-CMs as suitable in vitro models for investigating CYP2J2 in drug metabolism and cardiotoxicity. We first performed the genotyping and confirmed the presence of wild-type CYP2J2∗1/∗1 alleles in wild-type hESCs. Our optimized cardiomyocyte differentiation protocols yielded virtually pure (93.3% ± 6.8%) hESC-CMs, which exhibited P450 epoxygenase mRNA-expression profiles consistent with human cardiomyocytes, with CYP2J2 as the dominant isozyme and minor contributions from CYP2C8 and CYP2C9. By employing a CYP2J2-selective fluorescent substrate, ER-BnXPI, and astemizole as probe substrates, CYP2J2-mediated demethylation of both substrates exhibited typical Michaelis-Menten kinetics, which confirms functional CYP2J2 activities in vitro. Additionally, we demonstrated the capacity of CYP2J2 for arachidonic acid epoxidation, validating its ability to metabolize polyunsaturated fatty acid substrates. Finally, CYP2J2 activity in hESC-CMs was significantly inhibited by danazol and dronedarone, which are established CYP2J2 inhibitors known to cause cardiotoxicity. Ultimately, our study sheds novel insights on hESC-CMs as a suitable model for investigating CYP2J2-mediated metabolism and its inhibition in vitro. SIGNIFICANCE STATEMENT: H7 human embryonic stem cell-derived cardiomyocytes (hESC-CMs) were developed and validated as an in vitro model for investigating CYP2J2-mediated drug metabolism and its inhibition. By characterizing CYP2J2 transcriptional expression, catalytic activity, and inhibition response to established CYP2J2 inhibitors, our study confirmed functional CYP2J2 in hESC-CMs and ascertained that the model recapitulates the physiology of primary cardiomyocytes. This pioneering research highlights the potential of hESC-CMs in advancing our understanding of CYP2J2-mediated metabolism, its inhibition, and implications in drug-induced cardiotoxicity.

{"title":"Development of human embryonic stem cell-derived cardiomyocytes and application of fluorescence probe substrate for characterization of cytochrome P450 enzyme 2J2.","authors":"Xingyu Zhu, Yee Kiat Soh, Mingxin Wan, Jeremy Kah Sheng Pang, Wei Liang Leow, Chong Tian, Boon Seng Soh, Eric Chun Yong Chan","doi":"10.1016/j.dmd.2025.100053","DOIUrl":"https://doi.org/10.1016/j.dmd.2025.100053","url":null,"abstract":"<p><p>Cardiac cytochrome P450 2J2 (CYP2J2) plays a significant role in cardiovascular homeostasis due to its dual functions in drug metabolism and the epoxidation of polyunsaturated fatty acids. Additionally, the inhibition of CYP2J2 by xenobiotics has been linked to drug-induced cardiotoxicity, warranting further investigation of this critical enzyme in cardiac systems. Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) are physiologically relevant in vitro models that recapitulate relevant phenotypes important for cardiovascular research. However, no studies have so far characterized CYP2J2 expression and activities in these models. Here, we developed and validated H7 hESC-CMs as suitable in vitro models for investigating CYP2J2 in drug metabolism and cardiotoxicity. We first performed the genotyping and confirmed the presence of wild-type CYP2J2∗1/∗1 alleles in wild-type hESCs. Our optimized cardiomyocyte differentiation protocols yielded virtually pure (93.3% ± 6.8%) hESC-CMs, which exhibited P450 epoxygenase mRNA-expression profiles consistent with human cardiomyocytes, with CYP2J2 as the dominant isozyme and minor contributions from CYP2C8 and CYP2C9. By employing a CYP2J2-selective fluorescent substrate, ER-BnXPI, and astemizole as probe substrates, CYP2J2-mediated demethylation of both substrates exhibited typical Michaelis-Menten kinetics, which confirms functional CYP2J2 activities in vitro. Additionally, we demonstrated the capacity of CYP2J2 for arachidonic acid epoxidation, validating its ability to metabolize polyunsaturated fatty acid substrates. Finally, CYP2J2 activity in hESC-CMs was significantly inhibited by danazol and dronedarone, which are established CYP2J2 inhibitors known to cause cardiotoxicity. Ultimately, our study sheds novel insights on hESC-CMs as a suitable model for investigating CYP2J2-mediated metabolism and its inhibition in vitro. SIGNIFICANCE STATEMENT: H7 human embryonic stem cell-derived cardiomyocytes (hESC-CMs) were developed and validated as an in vitro model for investigating CYP2J2-mediated drug metabolism and its inhibition. By characterizing CYP2J2 transcriptional expression, catalytic activity, and inhibition response to established CYP2J2 inhibitors, our study confirmed functional CYP2J2 in hESC-CMs and ascertained that the model recapitulates the physiology of primary cardiomyocytes. This pioneering research highlights the potential of hESC-CMs in advancing our understanding of CYP2J2-mediated metabolism, its inhibition, and implications in drug-induced cardiotoxicity.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 4","pages":"100053"},"PeriodicalIF":4.4,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143742636","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}
引用次数: 0
Impact of the loss of slc43a3 on 6-mercaptopurine absorption and tissue distribution in mice.
IF 4.4 3区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-03-03 DOI: 10.1016/j.dmd.2025.100054
Aaron L Sayler, Hannah Dean, James R Hammond

6-Mercaptopurine (6-MP) is a nucleobase analog used in the therapy of acute lymphoblastic leukemia and inflammatory bowel disease. It is associated with numerous side effects including myelotoxicity, hepatotoxicity, and gastrointestinal complications, which can lead to patient adherence issues or discontinuation of treatment. This is further complicated by the wide variability in plasma levels of 6-MP and the therapeutic response to a standard dose. Although a number of enzyme polymorphisms have been linked to therapeutic response, it is unclear what factors underlie the variability in plasma levels. We have established that SLC43A3-encoded equilibrative nucleobase transporter 1 mediates the transport of 6-MP into cells in both mice and humans. To determine whether this transporter is critical for 6-MP absorption and biodistribution, we examined the effect of the genetic deletion of slc43a3 in mice on the absorption and tissue distribution of orally administered 6-MP. A high-performance liquid chromatography method was developed to measure tissue levels of 6-MP and its key metabolites, 6-methylmercaptoprine, 6-thiourate, and 6-thioguanine nucleotides. The results of this study show that loss of slc43a3 dramatically reduces the absorption of 6-MP from the gastrointestinal tract and attenuates the levels achieved in peripheral tissues. Furthermore, the loss of slc43a3 decreases the tissue:blood concentration ratios of 6-MP and its metabolites, particularly in those tissues that show high levels of expression of slc43a3, such as the heart and lungs. Therefore, it is possible that differences in SLC43A3 expression in humans may contribute to the variability seen in 6-MP plasma levels and therapeutic response. SIGNIFICANCE STATEMENT: The loss of slc43a3 in mice dramatically reduces the absorption and the biodistribution of the chemotherapeutic drug 6-mercaptopurine. These data suggest that variations in SLC43A3 expression in humans may contribute to the variability in plasma levels that have been reported when using this drug therapeutically.

6-巯基嘌呤(6-MP)是一种核碱基类似物,用于治疗急性淋巴细胞白血病和炎症性肠病。它有许多副作用,包括骨髓毒性、肝毒性和胃肠道并发症,这些副作用可能导致患者不能坚持治疗或中断治疗。由于 6-MP 的血浆水平和对标准剂量的治疗反应存在很大差异,这使得治疗变得更加复杂。虽然一些酶的多态性与治疗反应有关,但目前还不清楚是什么因素导致了血浆水平的变化。我们已经确定,由 SLC43A3 编码的平衡核碱基转运体 1 在小鼠和人体内都能介导 6-MP 向细胞的转运。为了确定这种转运体是否对 6-MP 的吸收和生物分布至关重要,我们研究了小鼠基因缺失 slc43a3 对口服 6-MP 的吸收和组织分布的影响。我们开发了一种高效液相色谱法来测量组织中 6-MP及其主要代谢物 6-甲基巯基嘌呤、6-硫代硫酸酯和 6-硫鸟嘌呤核苷酸的含量。这项研究的结果表明,slc43a3 的缺失会大大减少胃肠道对 6-MP 的吸收,并降低外周组织中 6-MP 的含量。此外,slc43a3 的缺失还会降低 6-MP 及其代谢物在组织与血液中的浓度比,尤其是在那些高水平表达 slc43a3 的组织中,如心脏和肺部。因此,人体中 SLC43A3 表达的差异可能是导致 6-MP 血浆水平和治疗反应出现差异的原因。意义声明:小鼠体内 Slc43a3 的缺失大大降低了化疗药物 6-巯基嘌呤的吸收和生物分布。这些数据表明,人体内 SLC43A3 表达的变化可能是导致血浆中 6-巯基嘌呤水平变化的原因之一。
{"title":"Impact of the loss of slc43a3 on 6-mercaptopurine absorption and tissue distribution in mice.","authors":"Aaron L Sayler, Hannah Dean, James R Hammond","doi":"10.1016/j.dmd.2025.100054","DOIUrl":"https://doi.org/10.1016/j.dmd.2025.100054","url":null,"abstract":"<p><p>6-Mercaptopurine (6-MP) is a nucleobase analog used in the therapy of acute lymphoblastic leukemia and inflammatory bowel disease. It is associated with numerous side effects including myelotoxicity, hepatotoxicity, and gastrointestinal complications, which can lead to patient adherence issues or discontinuation of treatment. This is further complicated by the wide variability in plasma levels of 6-MP and the therapeutic response to a standard dose. Although a number of enzyme polymorphisms have been linked to therapeutic response, it is unclear what factors underlie the variability in plasma levels. We have established that SLC43A3-encoded equilibrative nucleobase transporter 1 mediates the transport of 6-MP into cells in both mice and humans. To determine whether this transporter is critical for 6-MP absorption and biodistribution, we examined the effect of the genetic deletion of slc43a3 in mice on the absorption and tissue distribution of orally administered 6-MP. A high-performance liquid chromatography method was developed to measure tissue levels of 6-MP and its key metabolites, 6-methylmercaptoprine, 6-thiourate, and 6-thioguanine nucleotides. The results of this study show that loss of slc43a3 dramatically reduces the absorption of 6-MP from the gastrointestinal tract and attenuates the levels achieved in peripheral tissues. Furthermore, the loss of slc43a3 decreases the tissue:blood concentration ratios of 6-MP and its metabolites, particularly in those tissues that show high levels of expression of slc43a3, such as the heart and lungs. Therefore, it is possible that differences in SLC43A3 expression in humans may contribute to the variability seen in 6-MP plasma levels and therapeutic response. SIGNIFICANCE STATEMENT: The loss of slc43a3 in mice dramatically reduces the absorption and the biodistribution of the chemotherapeutic drug 6-mercaptopurine. These data suggest that variations in SLC43A3 expression in humans may contribute to the variability in plasma levels that have been reported when using this drug therapeutically.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 4","pages":"100054"},"PeriodicalIF":4.4,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143708999","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}
引用次数: 0
Detection of antidrug antibodies against antibody-drug conjugates by solid-phase extraction with acid dissociation in cynomolgus monkey serum.
IF 4.4 3区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-03-01 Epub Date: 2025-01-21 DOI: 10.1016/j.dmd.2025.100039
Susan Chen, Konstantin Piatkov, Linlin Dong, Hiroshi Sugimoto

The formation of antidrug antibodies (ADAs) against antibody-drug conjugates (ADCs) can trigger a humoral immune response and change drug exposure. Although the immunogenicity assessment of an ADC drug in nonclinical nonhuman primates may not directly translate to potential immunogenicity in humans, the nonclinical ADA assay facilitates understanding the pharmacokinetic profiles of biotherapeutics. The immune response against the human IgG4 monoclonal antibody-based ADC was suspected in cynomolgus monkey serum after intravenous administration at 1.5 mg/kg. However, the conventional bridging format ADA assay presented unique challenges for the ADC molecules due to the interaction of ADC-based capture and detection reagents, which generated high background noise. Solid-phase extraction with acid dissociation (SPEAD) sample treatment allowed the selective ADA transfer to a second plate for detection while avoiding the interaction between the capture and detection reagents. The signal-to-noise ratio in the ADA assay for ADCs was notably improved with SPEAD sample treatment compared with the results from the bridging assay. Importantly, the rapid drug clearance of the ADC molecules at the later time points was well correlated with the signal-to-noise ratio of the ADA assay in monkey serum, suggesting the validity of the results. Hence, we demonstrated the utility of the SPEAD sample treatment to mitigate the critical reagent interaction that triggered the unexpectedly high background in the ADA assay. SIGNIFICANCE STATEMENT: A fit-for-purpose antidrug antibody screening assay for the human IgG4 monoclonal antibody-based antibody-drug conjugate (ADC) molecule by solid-phase extraction with acid dissociation was developed to mitigate the high background noise due to the interaction of capture and detection ADCs. A positive antidrug antibody signal was observed in the monkey serum sample, which is in line with the significant decrease in the plasma concentration of ADCs at the later time points.

{"title":"Detection of antidrug antibodies against antibody-drug conjugates by solid-phase extraction with acid dissociation in cynomolgus monkey serum.","authors":"Susan Chen, Konstantin Piatkov, Linlin Dong, Hiroshi Sugimoto","doi":"10.1016/j.dmd.2025.100039","DOIUrl":"10.1016/j.dmd.2025.100039","url":null,"abstract":"<p><p>The formation of antidrug antibodies (ADAs) against antibody-drug conjugates (ADCs) can trigger a humoral immune response and change drug exposure. Although the immunogenicity assessment of an ADC drug in nonclinical nonhuman primates may not directly translate to potential immunogenicity in humans, the nonclinical ADA assay facilitates understanding the pharmacokinetic profiles of biotherapeutics. The immune response against the human IgG4 monoclonal antibody-based ADC was suspected in cynomolgus monkey serum after intravenous administration at 1.5 mg/kg. However, the conventional bridging format ADA assay presented unique challenges for the ADC molecules due to the interaction of ADC-based capture and detection reagents, which generated high background noise. Solid-phase extraction with acid dissociation (SPEAD) sample treatment allowed the selective ADA transfer to a second plate for detection while avoiding the interaction between the capture and detection reagents. The signal-to-noise ratio in the ADA assay for ADCs was notably improved with SPEAD sample treatment compared with the results from the bridging assay. Importantly, the rapid drug clearance of the ADC molecules at the later time points was well correlated with the signal-to-noise ratio of the ADA assay in monkey serum, suggesting the validity of the results. Hence, we demonstrated the utility of the SPEAD sample treatment to mitigate the critical reagent interaction that triggered the unexpectedly high background in the ADA assay. SIGNIFICANCE STATEMENT: A fit-for-purpose antidrug antibody screening assay for the human IgG4 monoclonal antibody-based antibody-drug conjugate (ADC) molecule by solid-phase extraction with acid dissociation was developed to mitigate the high background noise due to the interaction of capture and detection ADCs. A positive antidrug antibody signal was observed in the monkey serum sample, which is in line with the significant decrease in the plasma concentration of ADCs at the later time points.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 3","pages":"100039"},"PeriodicalIF":4.4,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143556131","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}
引用次数: 0
A review of physiologically based pharmacokinetic modeling of renal drug disposition.
IF 4.4 3区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-03-01 Epub Date: 2025-01-21 DOI: 10.1016/j.dmd.2025.100042
Weize Huang, Christine Bowman, Mengyue Yin, Lyrialle W Han, Yue Winnie Wen, Sara Kim Ahn, Yuan Chen

The human kidney is a critical organ for the elimination of numerous drugs and metabolites. The mechanisms of renal drug handling are manifold including unbound filtration, transporter-mediated active secretion, bidirectional passive diffusion, and occasionally active reabsorption and renal metabolism. These mechanisms collectively dictate the fate of drugs at various spatiotemporal points as drug molecules travel through the renal vasculature, tubules, and cells, posing a significant challenge in accurately describing and predicting renal drug disposition. Toward this end, a physiologically based kidney model serves as a promising tool to combine the anatomical and physiological features of the kidney (eg, tubular flow rate, pH, and transporter expression) with the unique properties of drugs (eg, protein binding, lipophilicity, ionization, and transporter substrate) to capture the dynamic system-drug interactions. Despite the exciting progress over the past several decades, physiologically based pharmacokinetic modeling has overall been predominantly used to predict intestinal absorption and hepatic drug-drug interaction. In comparison, pharmacokinetic modeling of renal drug handling has been underappreciated. In this review, we first provide an overview of kidney function and physiology, renal clearance mechanisms, and the evolutionary history of the physiologically based mechanistic kidney model. We then summarize the recent efforts spent in different areas of kidney model application, particularly: (1) renal transporter-mediated drug-drug interaction, (2) disease effect from both renal and hepatic impairment, and (3) model applications across the lifespan (eg, pediatrics and geriatrics). Finally, we identify remaining knowledge gaps, future directions, and potential model utilities. SIGNIFICANCE STATEMENT: This review summarizes pharmacokinetic model case studies that are related to renal drug disposition, illustrating the current framework of modeling renal drug handling, highlighting knowledge gaps in predicting renal transporter-mediated drug-drug interactions, and modeling the effects of disease and age on renal drug handling. A discussion on robust model validation and areas for future directions is also provided.

{"title":"A review of physiologically based pharmacokinetic modeling of renal drug disposition.","authors":"Weize Huang, Christine Bowman, Mengyue Yin, Lyrialle W Han, Yue Winnie Wen, Sara Kim Ahn, Yuan Chen","doi":"10.1016/j.dmd.2025.100042","DOIUrl":"10.1016/j.dmd.2025.100042","url":null,"abstract":"<p><p>The human kidney is a critical organ for the elimination of numerous drugs and metabolites. The mechanisms of renal drug handling are manifold including unbound filtration, transporter-mediated active secretion, bidirectional passive diffusion, and occasionally active reabsorption and renal metabolism. These mechanisms collectively dictate the fate of drugs at various spatiotemporal points as drug molecules travel through the renal vasculature, tubules, and cells, posing a significant challenge in accurately describing and predicting renal drug disposition. Toward this end, a physiologically based kidney model serves as a promising tool to combine the anatomical and physiological features of the kidney (eg, tubular flow rate, pH, and transporter expression) with the unique properties of drugs (eg, protein binding, lipophilicity, ionization, and transporter substrate) to capture the dynamic system-drug interactions. Despite the exciting progress over the past several decades, physiologically based pharmacokinetic modeling has overall been predominantly used to predict intestinal absorption and hepatic drug-drug interaction. In comparison, pharmacokinetic modeling of renal drug handling has been underappreciated. In this review, we first provide an overview of kidney function and physiology, renal clearance mechanisms, and the evolutionary history of the physiologically based mechanistic kidney model. We then summarize the recent efforts spent in different areas of kidney model application, particularly: (1) renal transporter-mediated drug-drug interaction, (2) disease effect from both renal and hepatic impairment, and (3) model applications across the lifespan (eg, pediatrics and geriatrics). Finally, we identify remaining knowledge gaps, future directions, and potential model utilities. SIGNIFICANCE STATEMENT: This review summarizes pharmacokinetic model case studies that are related to renal drug disposition, illustrating the current framework of modeling renal drug handling, highlighting knowledge gaps in predicting renal transporter-mediated drug-drug interactions, and modeling the effects of disease and age on renal drug handling. A discussion on robust model validation and areas for future directions is also provided.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 3","pages":"100042"},"PeriodicalIF":4.4,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143572441","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}
引用次数: 0
期刊
Drug Metabolism and Disposition
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
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
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1