Drug Metabolism and Disposition最新文献

筛选
英文 中文
Functional studies on the cytochrome P450 splice variants CYP4F3A and CYP4F3B unveil the basis for their distinct physiological functions. 细胞色素P450剪接变异体CYP4F3A和CYP4F3B的功能研究揭示了它们不同生理功能的基础。
IF 4 3区 医学
Drug Metabolism and Disposition Pub Date : 2025-09-29 DOI: 10.1016/j.dmd.2025.100176
Brisa Caroline Alves Chagas, Bjoern Brixius, Somayeh Pirhadi, Adriana Mirtchev, Sutapa Ray, David R Koes, Simone Brixius-Anderko
{"title":"Functional studies on the cytochrome P450 splice variants CYP4F3A and CYP4F3B unveil the basis for their distinct physiological functions.","authors":"Brisa Caroline Alves Chagas, Bjoern Brixius, Somayeh Pirhadi, Adriana Mirtchev, Sutapa Ray, David R Koes, Simone Brixius-Anderko","doi":"10.1016/j.dmd.2025.100176","DOIUrl":"https://doi.org/10.1016/j.dmd.2025.100176","url":null,"abstract":"<p><p>The cytochrome P450 (P450) 4F family (CYP4F) are fatty acid ⍵-hydroxylases that catalyze the insertion of a hydroxyl group at the terminal carbon. The enzymes CYP4F3A and CYP4F3B are special cases among all other human P450 enzymes because they are derived from the same gene. The CYP4F3 gene undergoes alternative splicing, resulting in the 2 distinct enzymes. CYP4F3A is exclusively expressed in monocytes and deactivates leukotriene B4 as part of the anti-inflammatory response. Conversely, CYP4F3B is expressed in the liver and kidney where its major function is the production of the potent lipid mediator 20-hydroxyeicosatetraenoic acid from arachidonic acid. Despite these differences, they share a 93% amino acid sequence identity because of their shared gene locus. Both CYP4F3A and CYP4F3B are potential therapeutic targets for autoimmune disorders, cardiovascular diseases, and cancer. Because there is a significant gap in understanding enzyme function, their use as therapeutic targets has not been realized yet. To our knowledge, we present the first protocol for the generation of functional recombinant CYP4F3A and CYP4F3B to high purity. Catalytic assays with arachidonic acid and leukotriene B4 reveal a distinct substrate preference of both enzymes, which confirm their distinct body functions. Spectral analysis confirmed a different binding mode of arachidonic acid to the splice variants with a differential interaction with the respective active site. In addition, we tested the inhibitory effect of the CYP4 pan inhibitor HET0016 on both variants. In conclusion, we successfully implemented a robust protocol for the production of recombinant CYP4F3A and CYP4F3B, which paves the way for more in-depth mechanistic and structural studies and future directed drug design. SIGNIFICANCE STATEMENT: The splice variants CYP4F3A and CYP4F3B originate from the same gene but assume different functions in the human body. However, in-depth structural and functional studies are missing owing to the lack of robust protein expression protocols. In this study, we achieved the first generation of recombinant enzyme and conducted functional studies with fatty acid substrates and drugs, paving a way to a deeper understanding of these fascinating enzymes.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 11","pages":"100176"},"PeriodicalIF":4.0,"publicationDate":"2025-09-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145354150","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
Multispecies minimal physiologically based pharmacokinetic-pharmacodynamic model of antisense oligonucleotides for central nervous system disorders. 中枢神经系统疾病的反义寡核苷酸多物种最小生理药代动力学-药效学模型。
IF 4 3区 医学
Drug Metabolism and Disposition Pub Date : 2025-09-24 DOI: 10.1016/j.dmd.2025.100167
Devam A Desai, Mfonabasi E Ette, Dhaval K Shah, Donald E Mager
{"title":"Multispecies minimal physiologically based pharmacokinetic-pharmacodynamic model of antisense oligonucleotides for central nervous system disorders.","authors":"Devam A Desai, Mfonabasi E Ette, Dhaval K Shah, Donald E Mager","doi":"10.1016/j.dmd.2025.100167","DOIUrl":"https://doi.org/10.1016/j.dmd.2025.100167","url":null,"abstract":"<p><p>Antisense oligonucleotides (ASOs) are selective small biological drugs used to decrease targeted proteins by suppressing mRNA expression. In this study, a quantitative framework was developed to characterize the disposition and effects of such drugs in the central nervous system across species to facilitate the translation of preclinical pharmacology to the clinic. A minimal physiologically based pharmacokinetic (PK)-pharmacodynamic (PD) model for ASOs was developed using published drug and species-specific physiological information, including PK of nusinersen and preclinical PK and mRNA expression for an investigational compound targeting glutamate receptor subunit 1. The model describes relevant pharmacological processes, including (1) clathrin-dependent/independent endocytosis, (2) exocytosis, (3) exonuclease metabolism, (4) macropinocytosis, and (5) knockdown of the targeted protein. Most physiological values were obtained from literature, and drug-specific parameters were estimated. The model captured PK data in preclinical species (mice, rats, and monkeys), infants, and pediatric subjects from phase 1 and 2 studies. Renal clearances were fixed to 2.07, 25.2, 170.7, and 405 mL/h for mice, rats, monkeys, and humans, which were based on prior published values for oligonucleotides. Glutamate receptor subunit 1 mRNA and protein expression in rats were well characterized using a precursor-dependent indirect response model assuming maximal inhibition (I<sub>max</sub>) set to 1. Overall, the biodistribution of 2 ASOs across species were characterized by implementing allometric scaling and minimal physiologically based PK concepts. The final model provides insights into the role of specific disposition processes in controlling ASO PK-PD properties in the central nervous system. SIGNIFICANCE STATEMENT: Antisense oligonucleotides enable central nervous system-targeted gene therapy and precision medicine. Using a translational multispecies minimal physiologically based PK-PD model, drug- and system-specific factors were identified that influence the biodistribution of antisense oligonucleotides in preclinical species and humans. The final model can readily translate preclinical data to anticipate human drug exposures in the brain and plasma, help in lead and backup compound selection, project first-in-human dose levels, interpret early human PK-PD data, and facilitate the identification of recommended phase 2 doses.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 11","pages":"100167"},"PeriodicalIF":4.0,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145307096","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
Quetiapine competitively inhibits aldehyde oxidase-mediated reduction. 喹硫平竞争性地抑制醛氧化酶介导的还原。
IF 4 3区 医学
Drug Metabolism and Disposition Pub Date : 2025-09-24 DOI: 10.1016/j.dmd.2025.100169
Hinata Ueda, Shuho Asano, Katsuya Narumi, Ryoichi Aoyagi, Keisuke Okamoto, Masaki Kobayashi
{"title":"Quetiapine competitively inhibits aldehyde oxidase-mediated reduction.","authors":"Hinata Ueda, Shuho Asano, Katsuya Narumi, Ryoichi Aoyagi, Keisuke Okamoto, Masaki Kobayashi","doi":"10.1016/j.dmd.2025.100169","DOIUrl":"https://doi.org/10.1016/j.dmd.2025.100169","url":null,"abstract":"<p><p>Aldehyde oxidase (AOX) oxidizes nitrogen-containing heterocyclic drugs and reduces electron-deficient nitroaromatic drugs. The aim of this study was to elucidate the mode of inhibition of benzothiazepines such as quetiapine and clozapine, which are known inhibitors of AOX, to predict drug-drug interactions between AOX substrates and inhibitors. Quetiapine and its metabolites inhibited the oxidation and reduction activities of AOX (inhibitory effects: quetiapine ≈ norquetiapine > quetiapine sulfoxide > quetiapine carboxylic acid). The inhibition mode of quetiapine was noncompetitive for phthalazine oxidation (K<sub>i</sub>, 5.72 ± 0.88 μM) and competitive for flunitrazepam reduction (K<sub>i</sub>, 5.71 ± 0.34 μM). Although a mixed inhibition mode was indicated for the reduction of AOX by clozapine (K<sub>i</sub>, 30.91 ± 4.02 μM), the affinity for the enzyme-substrate complex was estimated to be lower than its affinity for the substrate-free enzyme. On the basis of these results, we expected that benzothiazepines would inhibit activity by becoming trapped in the pocket of AOX, where the electron donor resides. Quetiapine and its metabolites did not inhibit xanthine oxidase activity, and it is assumed that there are significant structural differences in the sites where the reduction reactions of AOX and xanthine oxidase occur. To our knowledge, this is the first study to identify drugs that competitively inhibit the AOX-mediated reduction reactions. The affinities of the inhibitors, especially quetiapine, were higher than those of flunitrazepam used in this study. When evaluating the combined effects of competitive inhibitors on substrate drugs, attention should be paid to the concentrations of both the substrate and the inhibitor. SIGNIFICANCE STATEMENT: Quetiapine inhibited the oxidative reaction of aldehyde oxidase noncompetitively and the reductive reaction competitively, suggesting that benzothiazepines tend to bind to the reductive pocket of aldehyde oxidase.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 11","pages":"100169"},"PeriodicalIF":4.0,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145307222","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
Activation of pregnane X receptor-organic anion transporting polypeptide 1a/b /P-glycoprotein/multidrug resistance protein 2 axis mediates the accelerated blood and liver clearance of PEGylated liposomes. 孕烷X受体-有机阴离子转运多肽1a/b / p -糖蛋白/多药耐药蛋白2轴的激活介导了聚乙二醇化脂质体血液和肝脏的加速清除。
IF 4 3区 医学
Drug Metabolism and Disposition Pub Date : 2025-09-24 DOI: 10.1016/j.dmd.2025.100172
Jianquan Pan, Cheng Zhang, Huiyu Lu, Xue Zhang, Huiya Deng, Yunna Chen, Lei Wang, Sheng Zhang, Fengling Wang, Weidong Chen
{"title":"Activation of pregnane X receptor-organic anion transporting polypeptide 1a/b /P-glycoprotein/multidrug resistance protein 2 axis mediates the accelerated blood and liver clearance of PEGylated liposomes.","authors":"Jianquan Pan, Cheng Zhang, Huiyu Lu, Xue Zhang, Huiya Deng, Yunna Chen, Lei Wang, Sheng Zhang, Fengling Wang, Weidong Chen","doi":"10.1016/j.dmd.2025.100172","DOIUrl":"https://doi.org/10.1016/j.dmd.2025.100172","url":null,"abstract":"<p><p>Previous studies revealed that pregnane X receptor (PXR) was involved in the \"accelerated blood clearance (ABC)\" phenomenon induced by repeated injections of PEGylated liposomes, and observed the quickly reduced hepatic accumulation of the second dose in rats at 12 hours after the last injection, compared with the increased hepatic accumulation characteristic of the ABC phenomenon. Among the downstream target genes of PXR, organic anion transporting polypeptide 1a/b (Oatp1a/b), P-glycoprotein (P-gp), and multidrug resistance protein 2 (Mrp2) are responsible for regulating the absorption, distribution, and efflux processes of drugs. This study aimed to investigate the molecular mechanisms behind the accelerated blood and liver clearance of the second dose of PEGylated liposomes from the perspective of PXR-regulated transporters. This study demonstrated that PXR activation promoted the blood clearance and liver clearance of the second dose of PEGylated liposomes in rats via the upregulation of Oatp1a4/1b2 and P-gp/Mrp2, respectively. The long-circulation characteristic and intrahepatic disposition fate of PEGylated liposomes could be reconstructed by inhibition of the PXR-Oatp1a/b/P-gp/Mrp2 axis. Additionally, we found that the repeated injections of PEGylated liposomes could cause significant transporter-mediated drug-drug interactions in combination administration regimens due to the activation of Oatp1a/b and P-gp/Mrp2. This study identifies the PXR-Oatp1a/b/P-gp/Mrp2 axis as a critical molecular regulator of systemic clearance of PEGylated liposomes and provides a therapeutic strategy to mitigate the ABC phenomenon through targeted inhibition of the PXR-Oatp1a/b/P-gp/Mrp2 axis, which is instructive for promoting the development and translation of PEGylated nanoformulations and optimizing their dosing regimens. SIGNIFICANCE STATEMENT: This study reveals that pregnane X receptor activation mediates the accelerated blood and liver clearance of the second dose of PEGylated liposomes in rats via upregulating the organic anion transporting polypeptide 1a4/1b2 and P-glycoprotein/multidrug resistance protein 2, and that PEGylated liposomes can induce transporter-mediated drug-drug interactions in combination administration regimens. These findings advance the understanding for the mechanisms underlying the in vivo disposition fate of repeated injections of PEGylated liposomes, providing guidance and new clues for inhibiting accelerated blood clearance phenomenon and optimizing the dosing regimens of PEGylated formulations.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 11","pages":"100172"},"PeriodicalIF":4.0,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145312586","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
Alcohol dehydrogenase 4 and aldo-keto reductase 1A1 catalyze the oxidation of 4-hydroxytolbutamide, a metabolite of tolbutamide, in the human liver. 醇脱氢酶4和醛酮还原酶1A1在人肝脏中催化4-羟基甲苯磺丁酰胺的氧化,甲苯磺丁酰胺的代谢物。
IF 4 3区 医学
Drug Metabolism and Disposition Pub Date : 2025-09-24 DOI: 10.1016/j.dmd.2025.100173
Kazuya Shimomura, Rei Sato, Miyu Watanabe, Yuichiro Higuchi, Shotaro Uehara, Nao Yoneda, Masataka Nakano, Hiroshi Suemizu, Miki Nakajima, Tatsuki Fukami
{"title":"Alcohol dehydrogenase 4 and aldo-keto reductase 1A1 catalyze the oxidation of 4-hydroxytolbutamide, a metabolite of tolbutamide, in the human liver.","authors":"Kazuya Shimomura, Rei Sato, Miyu Watanabe, Yuichiro Higuchi, Shotaro Uehara, Nao Yoneda, Masataka Nakano, Hiroshi Suemizu, Miki Nakajima, Tatsuki Fukami","doi":"10.1016/j.dmd.2025.100173","DOIUrl":"https://doi.org/10.1016/j.dmd.2025.100173","url":null,"abstract":"<p><p>Tolbutamide is metabolized by cytochrome P450 2C9 into 4-hydroxytolbutamide (4-OH TB), which retains pharmacological activity. 4-OH TB is further oxidized to the inactive metabolite 4-carboxytolbutamide, likely via the intermediate tolbutamide aldehyde (4-CHO TB). Because the conversion of 4-OH TB to 4-CHO TB is considered the rate-limiting step, the enzyme(s) catalyzing this reaction may play a crucial role in determining drug efficacy. We aimed to identify the enzyme(s) responsible for this process in the human liver. 4-CHO TB was formed from 4-OH TB in human liver cytosol (HLC) in the presence of nicotinamide-adenine dinucleotide (NAD<sup>+</sup>). The relative activity factor approach revealed that this reaction was primarily attributed to alcohol dehydrogenase 4 (ADH4). Interestingly, 4-CHO TB was also formed in HLC in the presence of nicotinamide-adenine dinucleotide phosphate (NADP<sup>+</sup>), with a 1.6-fold higher intrinsic clearance than that of NAD<sup>+</sup>. Untargeted proteomic analysis revealed a significant correlation between aldo-keto reductase 1A1 (AKR1A1) protein levels and NADP<sup>+</sup>-dependent 4-CHO TB formation in 15 HLC samples (r = 0.627, P < .05). Recombinant AKR1A1 effectively catalyzed this reaction, contributing 92% of NADP<sup>+</sup>-dependent 4-CHO TB formation in HLC. Based on hepatic NAD<sup>+</sup> and NADP<sup>+</sup> concentrations and the expression levels of ADH4 and AKR1A1, AKR1A1 was estimated to contribute one-third of ADH4 to 4-CHO TB formation in the human liver. In conclusion, we demonstrated that ADH4 and AKR1A1 jointly mediate the oxidation of 4-OH TB to 4-CHO TB in the human liver, highlighting the novel role of AKR1A1 as an oxidase in drug metabolism. SIGNIFICANCE STATEMENT: This study identifies aldo-keto reductase 1A1 as a novel enzyme involved in the oxidation of 4-hydroxytolbutamide in the human liver. Alongside alcohol dehydrogenase 4, aldo-keto reductase 1A1 contributes to NADP<sup>+</sup>-dependent aldehyde formation, suggesting a previously unrecognized role in drug metabolism and variability in tolbutamide clearance.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 11","pages":"100173"},"PeriodicalIF":4.0,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145312574","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
Metabolic flux analysis of bile acid biosynthesis acidic pathway in HepG2 cells reveals CYP8B1 inhibition of azole antifungals. HepG2细胞胆汁酸生物合成酸途径代谢通量分析揭示了唑类抗真菌药物对CYP8B1的抑制作用。
IF 4 3区 医学
Drug Metabolism and Disposition Pub Date : 2025-09-23 DOI: 10.1016/j.dmd.2025.100168
Yixuan Wang, Tingting Yu, Xinjie Zhang, Yutong Wang, Lanlan Gui, Wushuang Zeng, Liang Huang, Ke Lan
{"title":"Metabolic flux analysis of bile acid biosynthesis acidic pathway in HepG2 cells reveals CYP8B1 inhibition of azole antifungals.","authors":"Yixuan Wang, Tingting Yu, Xinjie Zhang, Yutong Wang, Lanlan Gui, Wushuang Zeng, Liang Huang, Ke Lan","doi":"10.1016/j.dmd.2025.100168","DOIUrl":"https://doi.org/10.1016/j.dmd.2025.100168","url":null,"abstract":"<p><p>Sterol 12α-hydroxylase (CYP8B1) is a key regulator of bile acid (BA) homeostasis and an emerging therapeutic target for metabolic disorders. To address the challenge of cellular CYP8B1 inhibition characterization, this work developed a pharmacologically optimized HepG2 cells model using triiodothyronine-dexamethasone-bezafibrate (TDB) induction, which significantly enhances the 12α-hydroxylation activity along the acidic pathway of BA biosynthesis in HepG2 cells. Employing stable isotope tracing with apolipoprotein A1-solubilized 2,3,4-<sup>13</sup>C<sub>3</sub>-cholesterol, we established a liquid chromatography-mass spectrometry-based flux analysis platform to track de novo BA synthesis. Combined with a recombinant CYP8B1 assay, flux analysis revealed that CYP8B1 participates in cholic acid synthesis in HepG2 cells, typically via 12α-hydroxylation of 7α-hydroxy-3-oxo-4-cholestenoic acid and dihydroxycholestanoic acid. In TDB-HepG2 cells, azole antifungals exhibited differentiated inhibition of 12α-hydroxylation activity, generally mirroring the enzymatic data. Econazole acted as a relatively selective CYP8B1 inhibitor with a cellular half-maximal inhibitory concentration of 0.31-0.45 μM, tioconazole and posaconazole dually inhibited CYP8B1 and sterol 27-hydroxylase (CYP27A1), itraconazole and voriconazole primarily inhibited CYP27A1, and fluconazole showed no activity toward either enzyme. This study provides the first direct evidence that CYP8B1 participates in cholic acid synthesis via the acidic pathway and establishes a high-throughput cellular platform for screening CYP8B1 inhibitors, revealing azoles as effective modulators of this pathway. SIGNIFICANCE STATEMENT: Optimized HepG2 model using a <sup>13</sup>C<sub>3</sub>-cholesterol flux assay provides direct evidence that CYP8B1 participates in cholic acid biosynthesis via the acidic pathway and establishes a high-throughput cellular platform for screening CYP8B1 inhibitors, revealing azoles as effective modulators of this pathway.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 11","pages":"100168"},"PeriodicalIF":4.0,"publicationDate":"2025-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145343797","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
The role of cytochrome P450 and gut microbiome in drug metabolism: Insights into Parkinson disease treatment. 细胞色素P450和肠道微生物组在药物代谢中的作用:对帕金森病治疗的见解。
IF 4 3区 医学
Drug Metabolism and Disposition Pub Date : 2025-09-19 DOI: 10.1016/j.dmd.2025.100166
Sindduja Muthukumar, Harysh Winster Suresh Babu, Anto George, Brij Mohan Maurya, Iyer Mahalaxmi, Mukesh Kumar Yadav, Dibbanti HariKrishna Reddy, Arvinder Wander, Arul Narayanasamy, Raja Ganesan, Vikas Lakhanpal, Balachandar Vellingiri
{"title":"The role of cytochrome P450 and gut microbiome in drug metabolism: Insights into Parkinson disease treatment.","authors":"Sindduja Muthukumar, Harysh Winster Suresh Babu, Anto George, Brij Mohan Maurya, Iyer Mahalaxmi, Mukesh Kumar Yadav, Dibbanti HariKrishna Reddy, Arvinder Wander, Arul Narayanasamy, Raja Ganesan, Vikas Lakhanpal, Balachandar Vellingiri","doi":"10.1016/j.dmd.2025.100166","DOIUrl":"https://doi.org/10.1016/j.dmd.2025.100166","url":null,"abstract":"<p><p>Parkinson disease (PD) is a complex neurodegenerative condition marked by progressive motor and nonmotor symptoms. Cytochrome P450 (P450) enzymes, notably those from the CYP1 and CYP2 families, are increasingly recognized as significant factors in the development of PD. This review examines the role of P450 enzymes in PD, covering genetic variations, copy number variations, and single nucleotide polymorphisms linked to PD pathogenicity. It also explores the regulatory mechanisms controlling P450 expression in PD and the influence of the gut microbiome and metabolites on P450 activity. Additionally, the review discusses how P450 enzymes metabolically activate drugs used to treat PD and investigates the intricate relationship between P450s and mitochondrial dysfunction. Finally, it underscores the therapeutic potential of targeting P450 enzymes for PD treatment. Understanding the diverse roles of P450 enzymes in PD may lead to innovative treatment approaches and personalized interventions for this challenging neurological disorder. SIGNIFICANCE STATEMENT: Cytochrome P450 (P450) enzymes significantly influence Parkinson disease (PD) development through their roles in drug metabolism and detoxification. Single nucleotide polymorphisms in P450 genes can alter enzyme activity, affecting PD susceptibility and progression. Gut microbiota modulates P450 function, impacting detoxification of PD-related toxins and influencing gut and blood-brain barrier integrity. Additionally, P450-mitochondrial interactions contribute to energy deficits and oxidative stress, exacerbating neurodegeneration in PD. Understanding these pathways may uncover novel therapeutic targets and personalized treatment strategies.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 11","pages":"100166"},"PeriodicalIF":4.0,"publicationDate":"2025-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145328517","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
Clinical pharmacokinetics and immunogenicity evaluation of a trophoblast cell surface antigen 2-targeted antibody-drug conjugate, FDA018, in patients with epithelial malignant solid tumors. 滋养层细胞表面抗原2靶向抗体-药物偶联物FDA018在上皮恶性实体瘤患者中的临床药代动力学和免疫原性评价
IF 4 3区 医学
Drug Metabolism and Disposition Pub Date : 2025-09-17 DOI: 10.1016/j.dmd.2025.100165
Minlu Cheng, Xianjing Li, Ya Li, Yiya Wang, Wenjia Li, Shuai Wang, Qinxin Song, Chang Shu, Li Ding
{"title":"Clinical pharmacokinetics and immunogenicity evaluation of a trophoblast cell surface antigen 2-targeted antibody-drug conjugate, FDA018, in patients with epithelial malignant solid tumors.","authors":"Minlu Cheng, Xianjing Li, Ya Li, Yiya Wang, Wenjia Li, Shuai Wang, Qinxin Song, Chang Shu, Li Ding","doi":"10.1016/j.dmd.2025.100165","DOIUrl":"https://doi.org/10.1016/j.dmd.2025.100165","url":null,"abstract":"<p><p>FDA018, a novel trophoblast cell surface antigen 2-targeted antibody-drug conjugate, is designed for treating trophoblast cell surface antigen 2-positive solid tumors, including triple-negative breast cancer, lung cancer, and gastric cancer. In this study, a single-center, open-label, dose-escalation clinical study was conducted to evaluate the pharmacokinetics (PK) and immunogenicity of FDA018 injection in patients with advanced or metastatic epithelial malignant solid tumors, using an \"accelerated titration\" design and a standard \"3+3\" design across 6 dose cohorts (2.5, 5, 7.5, 10, 12 and 15 mg/kg). PK profiles were investigated for a topoisomerase I inhibitor (SN-38)-conjugated antibody, total antibody, unconjugated SN-38, a glucuronate metabolite of SN-38 (SN-38G), and total SN-38. To evaluate the impact of immunogenicity on PK profiles of FDA018, immunogenicity was assessed with an electrochemiluminescence-based bridging immunoassay. PK results showed that the exposure of the SN-38-conjugated antibody increased linearly with dose escalation. The terminal half-life of the SN-38-conjugated antibody (about 30 hours) was significantly shorter than that of the total antibody (about 85 hours). The terminal half-life of the SN-38-conjugated antibody and total antibody appeared to be prolonged after multiple administrations. The exposure of unconjugated SN-38 accounted for approximately 1% of total SN-38, indicating that most SN-38 remained conjugated to the monoclonal antibody in the systemic circulation, and the off-target toxicity was controllable. The accumulation analysis results showed that total antibody accumulated after multiple administrations. For immunogenicity assessment, among 20 patients receiving doses from 7.5 to 15 mg/kg, 15% tested positive for anti-drug antibodies. Anti-drug antibody-positive results did not significantly affect the PK profiles of FDA018. In conclusion, FDA018 demonstrated promising PK and immunogenicity profiles and laid a solid foundation for further clinical research. SIGNIFICANCE STATEMENT: This study comprehensively evaluated the clinical pharmacokinetic and immunogenicity of the trophoblast cell surface antigen 2-targeted antibody-drug conjugate, FDA018, for the first time, to the authors' knowledge. Compared with the commercially available trophoblast cell surface antigen 2-targeted antibody-drug conjugate sacituzumab govitecan, FDA018 showed promising pharmacokinetic properties with higher exposure of conjugated antibody and lower exposure of unconjugated SN-38 in vivo, implying better efficacy and lower off-target toxicity.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 11","pages":"100165"},"PeriodicalIF":4.0,"publicationDate":"2025-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145279163","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
Structure and properties of an oxidative metabolite of 2-methoxy-N-[3-[4-[3-methyl-4-[(6-methyl-3-pyridinyl)oxy]anilino]-6-quinazolinyl]prop-2-enyl]acetamide generated by aldehyde oxidase. 醛氧化酶生成2-甲氧基- n -[3-[4-[3-甲基-4-[(6-甲基-3-吡啶基)氧]苯胺]-6-喹唑啉基]丙-2-烯基]乙酰胺的氧化代谢物的结构与性质
IF 4 3区 医学
Drug Metabolism and Disposition Pub Date : 2025-09-12 DOI: 10.1016/j.dmd.2025.100164
Kazuko Inoue, Motoharu Kakiki, Takafumi Komori
{"title":"Structure and properties of an oxidative metabolite of 2-methoxy-N-[3-[4-[3-methyl-4-[(6-methyl-3-pyridinyl)oxy]anilino]-6-quinazolinyl]prop-2-enyl]acetamide generated by aldehyde oxidase.","authors":"Kazuko Inoue, Motoharu Kakiki, Takafumi Komori","doi":"10.1016/j.dmd.2025.100164","DOIUrl":"https://doi.org/10.1016/j.dmd.2025.100164","url":null,"abstract":"<p><p>The drug candidate, 2-methoxy-N-[3-[4-[3-methyl-4-[(6-methyl-3-pyridinyl)oxy]anilino]-6-quinazolinyl]prop-2-enyl]acetamide (CP-724,714), was discontinued because of hepatotoxicity observed in clinical studies. CP-724,714 is a substrate of aldehyde oxidase (AO) found in the human liver cytosol. CP-724,714 metabolization by AO in cryopreserved human hepatocytes generates several oxidative metabolites, including (E)-N-(3-(2-hydroxy-4-(3-methyl-4-(6-methylpyridin-3-yloxy)phenylamino)quinazolin-6-yl)allyl)-2-methoxyacetamide (CP-724,714-AOM). However, the structure of CP-724,714-AOM has not been identified. Therefore, we aimed to identify the structure of CP-724,714-AOM, determine the propensity of CP-724,714 and CP-724,714-AOM for toxic effects, and understand the underlying hepatotoxic mechanism in humans. A synthesized oxidized CP-724,714, identified as CP-724,714-AOM, was consistent with the AO metabolite of CP-724,714 generated in the human liver cytosol. The enzymatic kinetic parameters of CP-724,714 were calculated as a K<sub>m</sub> value of 9.17 ± 0.70 μM and a V<sub>max</sub> value of 3.57 ± 0.10 pmol/min/mg of human liver cytosol proteins, assessed by the production of CP-724,714-AOM. CP-724,714 showed a weak inhibitory effect on the bile salt export pump without inhibiting multidrug resistance protein 2, whereas CP-724,714-AOM showed no inhibitory effect. The trapping assay showed that both compounds formed reactive metabolites in the human liver microsomes. Moreover, the inflammasome activation potentials of the compounds were observed in HepaRG cells, which can also generate CP-724,714-AOM from CP-724,714. Thus, our findings show that confirming AO susceptibility at an early stage of drug development is crucial for understanding the potential risks of AO metabolism in terms of pharmacokinetics and toxicity. SIGNIFICANCE STATEMENT: The structure of the aldehyde oxidase metabolite of 2-methoxy-N-[3-[4-[3-methyl-4-[(6-methyl-3-pyridinyl)oxy]anilino]-6-quinazolinyl]prop-2-enyl]acetamide (CP-724,714) was identified using a synthetic standard, and its kinetic parameters in the human liver cytosol were determined. Reactive metabolite formation and inflammasome activation by CP-724,714 and (E)-N-(3-(2-hydroxy-4-(3-methyl-4-(6-methylpyridin-3-yloxy)phenylamino)quinazolin-6-yl)allyl)-2-methoxyacetamide were observed.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 10","pages":"100164"},"PeriodicalIF":4.0,"publicationDate":"2025-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145212021","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
Structural characterization of in vivo and in vitro metabolites of bosutinib by liquid chromatography-tandem mass spectrometry, in combination with the in silico methodologies for toxicity and metabolism prediction. 采用液相色谱-串联质谱法对博舒替尼体内和体外代谢物进行结构表征,并结合计算机毒性和代谢预测方法。
IF 4 3区 医学
Drug Metabolism and Disposition Pub Date : 2025-09-11 DOI: 10.1016/j.dmd.2025.100161
Sowmya Chaganti, Nadeem Shaikh, Kavita Pimpre, Prateek Barik, Aditya Jadhav, Shrilekha Chilvery, Kalpana Talari, Chandraiah Godugu, Gananadhamu Samanthula
{"title":"Structural characterization of in vivo and in vitro metabolites of bosutinib by liquid chromatography-tandem mass spectrometry, in combination with the in silico methodologies for toxicity and metabolism prediction.","authors":"Sowmya Chaganti, Nadeem Shaikh, Kavita Pimpre, Prateek Barik, Aditya Jadhav, Shrilekha Chilvery, Kalpana Talari, Chandraiah Godugu, Gananadhamu Samanthula","doi":"10.1016/j.dmd.2025.100161","DOIUrl":"https://doi.org/10.1016/j.dmd.2025.100161","url":null,"abstract":"<p><p>Bosutinib monohydrate, a second-generation tyrosine kinase inhibitor, is primarily used to treat Philadelphia chromosome-positive chronic myelogenous leukemia. Pharmacokinetic studies in humans identified 3 metabolites of bosutinib: oxidative dechlorinated bosutinib, N-desmethylated bosutinib, and bosutinib N-oxide. Although a few metabolites have been reported clinically, a comprehensive understanding of bosutinib's metabolic fate is essential for optimizing its therapeutic use and minimizing risks. Therefore, the present study aimed to investigate the detailed metabolism of bosutinib using a combination of in vitro, in vivo, and in silico methods. In vitro experiments were conducted using liver microsomes and S9 fractions in the presence of suitable cofactors, whereas in vivo studies employed Sprague-Dawley rats in which bosutinib was administered as an oral suspension, followed by the collection of blood, urine, and feces at respective time points. The biological samples were analyzed using liquid chromatography-quadrupole-Orbitrap mass spectrometer. A total of 10 metabolites were identified, including 8 novel ones. The diverse metabolic reactions included oxidative O-dealkylation (B-M1, B-M2, B-M4, and B-M7), N-oxidation (B-M5), oxidative dechlorination (B-M2 and B-M3), N-dealkylation (B-M8 and B-M9), hydroxylation (B-M8), and glycine conjugation (B-M10). Interestingly, no metabolites were detected in the plasma, and the major metabolites, B-M3 (13.91%) and B-M9 (10.58%), were found predominantly in the feces. In silico predictions using Meteor Nexus matched with 6 of the experimentally observed metabolites. Toxicity and mutagenicity were further assessed using Deductive Estimation of Risk from Existing Knowledge Nexus and Structure Activity Relationship Analysis using Hypotheses Nexus, which indicated a potential mutagenic concern for B-M7. The integration of experimental and computational approaches in this work contributes significantly to understanding bosutinib's metabolic profile and can guide future strategies for its safe and effective clinical application. SIGNIFICANCE STATEMENT: This study provides an in-depth exploration of bosutinib's metabolic pathways using in vitro models and in vivo analysis of plasma, urine, and fecal samples. Prominently, in silico toxicity assessments indicated that B-M7 may pose mutagenic risks, emphasizing the need for further investigation.</p>","PeriodicalId":11309,"journal":{"name":"Drug Metabolism and Disposition","volume":"53 10","pages":"100161"},"PeriodicalIF":4.0,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145231645","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
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:604180095
Book学术官方微信