Pub Date : 2024-01-01Epub Date: 2023-11-03DOI: 10.1007/s13318-023-00862-4
Jairam Palamanda, Kelli J Glenn, Susan M Melnick
Background and objective: Cenobamate is an antiseizure medication (ASM) approved for treatment of focal epilepsy in adults. The objective of this study was to characterize the distribution, metabolism, and excretion of cenobamate in adult and pre- and postnatal rats, including pregnant and lactating females and nursing pups.
Methods: Distribution, metabolic, and excretion profiles were determined for 14C-labeled and unlabeled cenobamate using liquid scintillation counting, radiochromatography, LCMS, and LCMS/MS after oral or intravenous (IV) administration.
Results: Distribution of 14C-cenobamate-related material in adult male rats was widespread throughout the body, with nearly 1:1 tissue-to-plasma ratios observed for most tissues, including brain. Cenobamate administered to pregnant females was also transferred across the placental barrier into amniotic fluid and fetal plasma. Following administration to lactating F0 females, cenobamate was detected in breast milk and in plasma of nursing pups. 14C-cenobamate administered to adult male rats as a single oral dose was extensively metabolized with nine metabolites identified in urine and feces, including a principal dihydrodiol metabolite. Cenobamate was the principal drug-related material in rat plasma. Following a single dose of 14C-cenobamate to male and female rats, radioactivity was excreted equally into urine and feces, with mass balance achieved by 48 h postdose.
Conclusions: Distribution of cenobamate was widespread into many rat tissues, including brain, amniotic fluid, fetal plasma, breast milk, and breastfeeding rat pups. These distribution findings, along with the results of the metabolism and excretion studies, may help inform treatment decisions for patients with epilepsy being treated with cenobamate, including pregnant or nursing mothers.
{"title":"Distribution, Metabolism, and Excretion of Cenobamate in Adult, Fetal, Neonatal, and Lactating Rats.","authors":"Jairam Palamanda, Kelli J Glenn, Susan M Melnick","doi":"10.1007/s13318-023-00862-4","DOIUrl":"10.1007/s13318-023-00862-4","url":null,"abstract":"<p><strong>Background and objective: </strong>Cenobamate is an antiseizure medication (ASM) approved for treatment of focal epilepsy in adults. The objective of this study was to characterize the distribution, metabolism, and excretion of cenobamate in adult and pre- and postnatal rats, including pregnant and lactating females and nursing pups.</p><p><strong>Methods: </strong>Distribution, metabolic, and excretion profiles were determined for <sup>14</sup>C-labeled and unlabeled cenobamate using liquid scintillation counting, radiochromatography, LCMS, and LCMS/MS after oral or intravenous (IV) administration.</p><p><strong>Results: </strong>Distribution of <sup>14</sup>C-cenobamate-related material in adult male rats was widespread throughout the body, with nearly 1:1 tissue-to-plasma ratios observed for most tissues, including brain. Cenobamate administered to pregnant females was also transferred across the placental barrier into amniotic fluid and fetal plasma. Following administration to lactating F<sub>0</sub> females, cenobamate was detected in breast milk and in plasma of nursing pups. <sup>14</sup>C-cenobamate administered to adult male rats as a single oral dose was extensively metabolized with nine metabolites identified in urine and feces, including a principal dihydrodiol metabolite. Cenobamate was the principal drug-related material in rat plasma. Following a single dose of <sup>14</sup>C-cenobamate to male and female rats, radioactivity was excreted equally into urine and feces, with mass balance achieved by 48 h postdose.</p><p><strong>Conclusions: </strong>Distribution of cenobamate was widespread into many rat tissues, including brain, amniotic fluid, fetal plasma, breast milk, and breastfeeding rat pups. These distribution findings, along with the results of the metabolism and excretion studies, may help inform treatment decisions for patients with epilepsy being treated with cenobamate, including pregnant or nursing mothers.</p>","PeriodicalId":11939,"journal":{"name":"European Journal of Drug Metabolism and Pharmacokinetics","volume":" ","pages":"7-21"},"PeriodicalIF":1.9,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71434085","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-01-01Epub Date: 2023-12-01DOI: 10.1007/s13318-023-00871-3
Anastasiia Kalinina, Elena Grigorieva, Anna Smirnova, Dmitry Kazansky, Ludmila Khromykh
Background and objective: Cyclophilin A (CypA) is an isomerase that functions as a chaperone, housekeeping protein, and cyclosporine A (CsA) ligand. Secreted CypA is a proinflammatory factor, chemoattractant, immune regulator, and factor of antitumor immunity. Experimental data suggest clinical applications of recombinant human CypA (rhCypA) as a biotherapeutic for cancer immunotherapy, stimulation of tissue regeneration, treatment of brain pathologies, and as a supportive treatment for CsA-based therapies. The objective of this study is to analyze the pharmacokinetics of rhCypA in a mouse model.
Methods: rhCypA was isotope-labeled with 125I and injected intraperitoneally (i.p.) or subcutaneously (s/c) into female mice as a single dose of 100 μg per mouse, equivalent to the estimated first-in-human dose. Analysis of 125I-rhCypA biodistribution and excretion was performed by direct radiometry of the blood, viscera, and urine of mice 0.5-72 h following its administration.
Results: rhCypA showed rapid and even tissue-organ distribution, with the highest tropism (fT = 1.56) and accumulation (maximum concentration, Cmax = 137-167 μg/g) in the kidneys, its primary excretory organ. rhCypA showed the lowest tropism to the bone marrow and the brain (fT = 0.07) but the longest retention in these organs [mean retention time (MRT) = 25-28 h].
Conclusion: This study identified promising target organs for rhCypA's potential therapeutic effects. The mode of rhCypA accumulation and retention in organs could be primarily due to the expression of its receptors in them. For the first time, rhCypA was shown to cross the blood-brain barrier and accumulate in the brain. These rhCypA pharmacokinetic data could be extrapolated to humans as preliminary data for possible clinical trials.
{"title":"Pharmacokinetic Parameters of Recombinant Human Cyclophilin A in Mice.","authors":"Anastasiia Kalinina, Elena Grigorieva, Anna Smirnova, Dmitry Kazansky, Ludmila Khromykh","doi":"10.1007/s13318-023-00871-3","DOIUrl":"10.1007/s13318-023-00871-3","url":null,"abstract":"<p><strong>Background and objective: </strong>Cyclophilin A (CypA) is an isomerase that functions as a chaperone, housekeeping protein, and cyclosporine A (CsA) ligand. Secreted CypA is a proinflammatory factor, chemoattractant, immune regulator, and factor of antitumor immunity. Experimental data suggest clinical applications of recombinant human CypA (rhCypA) as a biotherapeutic for cancer immunotherapy, stimulation of tissue regeneration, treatment of brain pathologies, and as a supportive treatment for CsA-based therapies. The objective of this study is to analyze the pharmacokinetics of rhCypA in a mouse model.</p><p><strong>Methods: </strong>rhCypA was isotope-labeled with <sup>125</sup>I and injected intraperitoneally (i.p.) or subcutaneously (s/c) into female mice as a single dose of 100 μg per mouse, equivalent to the estimated first-in-human dose. Analysis of <sup>125</sup>I-rhCypA biodistribution and excretion was performed by direct radiometry of the blood, viscera, and urine of mice 0.5-72 h following its administration.</p><p><strong>Results: </strong>rhCypA showed rapid and even tissue-organ distribution, with the highest tropism (f<sub>T</sub> = 1.56) and accumulation (maximum concentration, C<sub>max</sub> = 137-167 μg/g) in the kidneys, its primary excretory organ. rhCypA showed the lowest tropism to the bone marrow and the brain (f<sub>T</sub> = 0.07) but the longest retention in these organs [mean retention time (MRT) = 25-28 h].</p><p><strong>Conclusion: </strong>This study identified promising target organs for rhCypA's potential therapeutic effects. The mode of rhCypA accumulation and retention in organs could be primarily due to the expression of its receptors in them. For the first time, rhCypA was shown to cross the blood-brain barrier and accumulate in the brain. These rhCypA pharmacokinetic data could be extrapolated to humans as preliminary data for possible clinical trials.</p>","PeriodicalId":11939,"journal":{"name":"European Journal of Drug Metabolism and Pharmacokinetics","volume":" ","pages":"57-69"},"PeriodicalIF":1.9,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138470059","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-12-19DOI: 10.1007/s13318-023-00875-z
Fang Zhang, Rui Wu, Yanfang Liu, Shu Dai, Xinyan Xue, Xiaohong Gong, Yunxia Li
Background and Objectives
Rhubarb anthraquinones contain five main components, that is, rhein, emodin, aloe-emodin, chrysophanol, and physcion, which demonstrate good therapeutic effects on nonalcoholic fatty liver disease (NAFLD). However, research on its pharmacokinetics in NAFLD remains lacking. This study aimed to investigate the pharmacokinetic differences of rhubarb anthraquinones in normal and NAFLD rats.
Methods
This study developed an NAFLD rat model by high-fat diet feeding for 6 weeks. Normal and NAFLD groups were orally administered different rhubarb anthraquinones doses (37.5, 75, and 150 mg/kg). The concentration of the rhein, emodin, aloe-emodin, chrysophanol, and physcion in plasma was determined by high-performance liquid chromatography–ultraviolet.
Results
The results revealed significant differences in pharmacokinetic behavior between normal and NAFLD rats. Compared with normal rats, NAFLD rats demonstrated significantly increased maximum plasma concentration (Cmax) and area under the plasma concentration–time curve (AUC0 → ∞) of rhubarb anthraquinones (P < 0.05), as well as significantly prolonged time to reach maximum plasma concentration (Tmax), terminal elimination half-life (t1/2), and mean residence time (MRT) of rhubarb anthraquinones (P < 0.05).
Conclusions
This study indicates significant differences in the pharmacokinetics of rhubarb anthraquinones between the physiological and NAFLD states of rats. Rhubarb anthraquinone demonstrated a longer retention time and slower absorption rate in NAFLD rats and exhibited higher bioavailability and peak concentration. This finding provides important information for guiding the clinical use of rhubarb anthraquinones under pathological conditions.
{"title":"Comparative Pharmacokinetic Study of Rhubarb Anthraquinones in Normal and Nonalcoholic Fatty Liver Disease Rats","authors":"Fang Zhang, Rui Wu, Yanfang Liu, Shu Dai, Xinyan Xue, Xiaohong Gong, Yunxia Li","doi":"10.1007/s13318-023-00875-z","DOIUrl":"https://doi.org/10.1007/s13318-023-00875-z","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Background and Objectives</h3><p>Rhubarb anthraquinones contain five main components, that is, rhein, emodin, aloe-emodin, chrysophanol, and physcion, which demonstrate good therapeutic effects on nonalcoholic fatty liver disease (NAFLD). However, research on its pharmacokinetics in NAFLD remains lacking. This study aimed to investigate the pharmacokinetic differences of rhubarb anthraquinones in normal and NAFLD rats.</p><h3 data-test=\"abstract-sub-heading\">Methods</h3><p>This study developed an NAFLD rat model by high-fat diet feeding for 6 weeks. Normal and NAFLD groups were orally administered different rhubarb anthraquinones doses (37.5, 75, and 150 mg/kg). The concentration of the rhein, emodin, aloe-emodin, chrysophanol, and physcion in plasma was determined by high-performance liquid chromatography–ultraviolet.</p><h3 data-test=\"abstract-sub-heading\">Results</h3><p>The results revealed significant differences in pharmacokinetic behavior between normal and NAFLD rats. Compared with normal rats, NAFLD rats demonstrated significantly increased maximum plasma concentration (<i>C</i><sub>max</sub>) and area under the plasma concentration–time curve (AUC<sub>0 → ∞</sub>) of rhubarb anthraquinones (<i>P</i> < 0.05), as well as significantly prolonged time to reach maximum plasma concentration (<i>T</i><sub>max</sub>), terminal elimination half-life (<i>t</i><sub>1/2</sub>), and mean residence time (MRT) of rhubarb anthraquinones (<i>P</i> < 0.05).</p><h3 data-test=\"abstract-sub-heading\">Conclusions</h3><p>This study indicates significant differences in the pharmacokinetics of rhubarb anthraquinones between the physiological and NAFLD states of rats. Rhubarb anthraquinone demonstrated a longer retention time and slower absorption rate in NAFLD rats and exhibited higher bioavailability and peak concentration. This finding provides important information for guiding the clinical use of rhubarb anthraquinones under pathological conditions.</p>","PeriodicalId":11939,"journal":{"name":"European Journal of Drug Metabolism and Pharmacokinetics","volume":"81 1","pages":""},"PeriodicalIF":1.9,"publicationDate":"2023-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138740898","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
There are some potential concerns about the currently marketed solid oral dosage forms of tramadol, including decreased adherence to immediate-release (IR) formulations due to the high number of doses taken each day and the slow rise in the blood tramadol concentration after administration of sustained-release (SR) formulations, which may not achieve a rapid analgesic effect. To overcome these potential concerns, a twice-daily double-layered tablet formulation of tramadol comprising IR and SR layers was developed. This article reports studies that assessed its physicochemical and pharmacokinetic properties.
Methods
Dissolution tests of five bilayer tablet formulations (designated tablets A–E) and pharmacokinetic studies of tablets A and B were conducted to investigate the appropriate ratio of the IR/SR layers in the double-layered tablet. Additionally, pharmacokinetic studies of three finished dosage formulations (tablets C–E) were performed in healthy adult males to investigate the effect of food intake on drug absorption.
Results
Adjusting the excipients and tramadol content in the IR and SR layers of tablets A–E altered their dissolution profiles in a manner that could be predicted based on their compositions. The IR layer was released within 15 min, and the SR layer was slowly released over 10 h. In the pharmacokinetic study, the time to maximum plasma concentration (tmax) of tramadol after administration of tablets A (IR:SR: 20:80 mg) and B (40:60 mg) was shorter than that of a commercially available SR tablet, and the half-life (t1/2) was longer than that of a commercially available IR tablet. For tablets C–E, administration after food did not affect the area under the concentration-time curve (AUC) or maximum drug concentration (Cmax) of tramadol, but the tmax was prolonged by about 1 h compared with administration in fasting conditions. The mean ± standard deviation tmax and t1/2 for tablet D (IR:SR: 35:65 mg) in the fasting condition was 1.09 ± 0.56 h and 7.82 ± 0.85 h, respectively. The respective values in the fed condition were 2.47 ± 1.06 h and 7.12 ± 0.85 h, respectively.
Conclusions
To address the potential concerns regarding existing formulations of tramadol, a twice-daily, extended-release bilayer formulation of tramadol consisting of an IR and SR layer was developed. Pharmacokinetic studies confirmed that the plasma tramadol concentration increased quickly after administration and was maintained over a long period of time.
背景和目的目前市场上销售的曲马多口服固体制剂存在一些潜在的问题,包括由于每天服用的剂量较多,服用速释(IR)制剂的依从性降低,以及服用缓释(SR)制剂后血中曲马多浓度上升缓慢,可能无法达到快速镇痛效果。为了克服这些潜在的问题,我们开发了一种由IR层和SR层组成的曲马多每日两次双层片剂。方法对五种双层片剂(指定为片剂 A-E)进行了溶解试验,并对片剂 A 和 B 进行了药代动力学研究,以探讨双层片剂中 IR 层/SR 层的适当比例。结果调整片剂 A-E 的 IR 层和 SR 层中的赋形剂和曲马多含量后,其溶解曲线发生了变化,而这种变化是可以根据其成分预测到的。在药代动力学研究中,服用 A 片(IR:SR:20:80 毫克)和 B 片(40:60 毫克)后曲马多达到最大血浆浓度(tmax)的时间短于市售的 SR 片,半衰期(t1/2)长于市售的 IR 片。对于 C-E 片剂,进食后给药不会影响曲马多的浓度曲线下面积(AUC)或最大药物浓度(Cmax),但与空腹给药相比,tmax 延长了约 1 小时。D 片(IR:SR:35:65 毫克)在空腹状态下的 tmax 和 t1/2 平均值(± 标准偏差)分别为 1.09 ± 0.56 小时和 7.82 ± 0.85 小时。结论为了解决现有曲马多制剂可能存在的问题,我们开发了一种曲马多的日服两次缓释双层制剂,由IR层和SR层组成。药代动力学研究证实,给药后血浆中曲马多的浓度会迅速升高并长期保持。
{"title":"Development, Physicochemical Characteristics and Pharmacokinetics of a New Sustained-Release Bilayer Tablet Formulation of Tramadol with an Immediate-Release Component for Twice-Daily Administration","authors":"Naoki Ishitsubo, Shinji Oguro, Hirotoshi Shimahashi, Masato Kawanishi, Takeshi Adachi, Kenji Mitsuda, Nobuyuki Ishibashi","doi":"10.1007/s13318-023-00865-1","DOIUrl":"https://doi.org/10.1007/s13318-023-00865-1","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Background and Objective</h3><p>There are some potential concerns about the currently marketed solid oral dosage forms of tramadol, including decreased adherence to immediate-release (IR) formulations due to the high number of doses taken each day and the slow rise in the blood tramadol concentration after administration of sustained-release (SR) formulations, which may not achieve a rapid analgesic effect. To overcome these potential concerns, a twice-daily double-layered tablet formulation of tramadol comprising IR and SR layers was developed. This article reports studies that assessed its physicochemical and pharmacokinetic properties.</p><h3 data-test=\"abstract-sub-heading\">Methods</h3><p>Dissolution tests of five bilayer tablet formulations (designated tablets A–E) and pharmacokinetic studies of tablets A and B were conducted to investigate the appropriate ratio of the IR/SR layers in the double-layered tablet. Additionally, pharmacokinetic studies of three finished dosage formulations (tablets C–E) were performed in healthy adult males to investigate the effect of food intake on drug absorption.</p><h3 data-test=\"abstract-sub-heading\">Results</h3><p>Adjusting the excipients and tramadol content in the IR and SR layers of tablets A–E altered their dissolution profiles in a manner that could be predicted based on their compositions. The IR layer was released within 15 min, and the SR layer was slowly released over 10 h. In the pharmacokinetic study, the time to maximum plasma concentration (<i>t</i><sub>max</sub>) of tramadol after administration of tablets A (IR:SR: 20:80 mg) and B (40:60 mg) was shorter than that of a commercially available SR tablet, and the half-life (<i>t</i><sub>1/2</sub>) was longer than that of a commercially available IR tablet. For tablets C–E, administration after food did not affect the area under the concentration-time curve (AUC) or maximum drug concentration (<i>C</i><sub>max</sub>) of tramadol, but the <i>t</i><sub>max</sub> was prolonged by about 1 h compared with administration in fasting conditions. The mean ± standard deviation <i>t</i><sub>max</sub> and <i>t</i><sub>1/2</sub> for tablet D (IR:SR: 35:65 mg) in the fasting condition was 1.09 ± 0.56 h and 7.82 ± 0.85 h, respectively. The respective values in the fed condition were 2.47 ± 1.06 h and 7.12 ± 0.85 h, respectively.</p><h3 data-test=\"abstract-sub-heading\">Conclusions</h3><p>To address the potential concerns regarding existing formulations of tramadol, a twice-daily, extended-release bilayer formulation of tramadol consisting of an IR and SR layer was developed. Pharmacokinetic studies confirmed that the plasma tramadol concentration increased quickly after administration and was maintained over a long period of time.</p>","PeriodicalId":11939,"journal":{"name":"European Journal of Drug Metabolism and Pharmacokinetics","volume":"19 1","pages":""},"PeriodicalIF":1.9,"publicationDate":"2023-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138555391","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-12-01Epub Date: 2023-06-14DOI: 10.1007/s12070-023-03943-7
Chee Chean Lim, Siew Wei Ling, Kumareysh Vijay Vijayan, Ming Jun Lee, Philip Rajan Devesayaham
Background: Acquired hemophilia A (AHA) is a rare blood disorder with high morbidity and even mortality as severe bleeding can occur in up to 90% of affected patients. Unlike congenital hemophilia which presents with intra-articular bleeding, acquired hemophilia causes bleeding into the skin, muscle, mucous membranes and soft tissues.
Case presentation: We report an unusual case of upper airway hematoma in a 61-year-old man who presented with acute onset dysphagia and shortness of breath. There were bruises on his neck and blood clots on the floor of mouth when he was examined. Endoscopic examination revealed an extensive laryngeal hematoma. A prolonged activated partial thromboplastin time (APTT) prompted us to investigate for factor VIII deficiency leading to the diagnosis of AHA. He recovered completely after a treatment regime instituted by the hematology team without suffering any grave debilitating events.
Conclusion: AHA with laryngeal hematoma is a rare condition with only a handful of cases reported. Although a life-threatening disease, it is easily reversed with early recognition and administration of medical therapy involving the hematology team.
{"title":"Acquired Hemophilia A: Rare Cause of Upper Airway Hematoma and a Literature Review.","authors":"Chee Chean Lim, Siew Wei Ling, Kumareysh Vijay Vijayan, Ming Jun Lee, Philip Rajan Devesayaham","doi":"10.1007/s12070-023-03943-7","DOIUrl":"10.1007/s12070-023-03943-7","url":null,"abstract":"<p><strong>Background: </strong>Acquired hemophilia A (AHA) is a rare blood disorder with high morbidity and even mortality as severe bleeding can occur in up to 90% of affected patients. Unlike congenital hemophilia which presents with intra-articular bleeding, acquired hemophilia causes bleeding into the skin, muscle, mucous membranes and soft tissues.</p><p><strong>Case presentation: </strong>We report an unusual case of upper airway hematoma in a 61-year-old man who presented with acute onset dysphagia and shortness of breath. There were bruises on his neck and blood clots on the floor of mouth when he was examined. Endoscopic examination revealed an extensive laryngeal hematoma. A prolonged activated partial thromboplastin time (APTT) prompted us to investigate for factor VIII deficiency leading to the diagnosis of AHA. He recovered completely after a treatment regime instituted by the hematology team without suffering any grave debilitating events.</p><p><strong>Conclusion: </strong>AHA with laryngeal hematoma is a rare condition with only a handful of cases reported. Although a life-threatening disease, it is easily reversed with early recognition and administration of medical therapy involving the hematology team.</p>","PeriodicalId":11939,"journal":{"name":"European Journal of Drug Metabolism and Pharmacokinetics","volume":"9 1","pages":"3886-3888"},"PeriodicalIF":0.6,"publicationDate":"2023-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10645700/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"73636068","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-11-01Epub Date: 2023-10-04DOI: 10.1007/s13318-023-00859-z
Xiang Chen, Guo Yu, Guo-Fu Li
{"title":"Use of Clearance Concepts to Simulate Impact of Interleukin-6 on Drug Elimination Governed by Cytochromes P450 3A4 and Glomerular Filtration Rate.","authors":"Xiang Chen, Guo Yu, Guo-Fu Li","doi":"10.1007/s13318-023-00859-z","DOIUrl":"10.1007/s13318-023-00859-z","url":null,"abstract":"","PeriodicalId":11939,"journal":{"name":"European Journal of Drug Metabolism and Pharmacokinetics","volume":" ","pages":"619-621"},"PeriodicalIF":1.9,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41104107","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-11-01Epub Date: 2023-09-26DOI: 10.1007/s13318-023-00854-4
Chun-Jui Huang, Chieh-Hua Lu, Kuang-Chung Shih
Background: In patients with kidney or hepatic diseases, an increment of circulating pasireotide is also expected. Therefore, this open-label, phase I study aimed to evaluate the pharmacokinetic profiles and safety of subcutaneous (SC) and long-acting release (LAR) intramuscular injections of pasireotide in male Taiwanese volunteers who are hyperendemic hepatitis B/C and chronic kidney disease (CKD).
Methods: A total of 45 male volunteers were randomized to receive one of nine treatment sequences, involving a single subcutaneous injection of 300, 600, or 900 μg pasireotide, a multiple SC injection of the same dosage of pasireotide [300, 600, or 900 μg, twice daily (b.i.d.) for 4 days and a single dose for 1 day], and a single dose of 20, 40, or 60 mg LAR pasireotide intramuscular injection. The pasireotide SC and LAR formulations were prepared and supplied to the study center by Novartis. Pharmacokinetic parameters were assessed from both formulations. All adverse events that occurred in participants throughout the study period, including abnormalities in fasting levels of glucose, insulin, and glucagon, as well as laboratory measurements and electrocardiograms, were recorded.
Results: Analysis of plasma concentration over time revealed a rapid absorption of pasireotide, with a maximal concentration at 0.5 h after SC injection(s) of pasireotide (300-900 µg). Following a single dose of pasireotide LAR (20-60 mg), a sustained release was observed following an initial increase on day 1, a plateau around day 20, and a decline over the next 7 weeks.
Conclusions: Both pasireotide formulations showed dose-proportional pharmacokinetics and 300-900 μg of SC pasireotide and 20-60 mg LAR pasireotide treatment showed favorable safety profiles and was well-tolerated when administered in male Taiwanese volunteers who are hyperendemic hepatitis B/C and CKD.
{"title":"Pharmacokinetics and Safety of Long-Acting Release Formulations of Pasireotide (SOM230) in a Male Population Who Are Hyperendemic Hepatitis B/C and Chronic Kidney Disease: An Open-Label, Phase I Study.","authors":"Chun-Jui Huang, Chieh-Hua Lu, Kuang-Chung Shih","doi":"10.1007/s13318-023-00854-4","DOIUrl":"10.1007/s13318-023-00854-4","url":null,"abstract":"<p><strong>Background: </strong>In patients with kidney or hepatic diseases, an increment of circulating pasireotide is also expected. Therefore, this open-label, phase I study aimed to evaluate the pharmacokinetic profiles and safety of subcutaneous (SC) and long-acting release (LAR) intramuscular injections of pasireotide in male Taiwanese volunteers who are hyperendemic hepatitis B/C and chronic kidney disease (CKD).</p><p><strong>Methods: </strong>A total of 45 male volunteers were randomized to receive one of nine treatment sequences, involving a single subcutaneous injection of 300, 600, or 900 μg pasireotide, a multiple SC injection of the same dosage of pasireotide [300, 600, or 900 μg, twice daily (b.i.d.) for 4 days and a single dose for 1 day], and a single dose of 20, 40, or 60 mg LAR pasireotide intramuscular injection. The pasireotide SC and LAR formulations were prepared and supplied to the study center by Novartis. Pharmacokinetic parameters were assessed from both formulations. All adverse events that occurred in participants throughout the study period, including abnormalities in fasting levels of glucose, insulin, and glucagon, as well as laboratory measurements and electrocardiograms, were recorded.</p><p><strong>Results: </strong>Analysis of plasma concentration over time revealed a rapid absorption of pasireotide, with a maximal concentration at 0.5 h after SC injection(s) of pasireotide (300-900 µg). Following a single dose of pasireotide LAR (20-60 mg), a sustained release was observed following an initial increase on day 1, a plateau around day 20, and a decline over the next 7 weeks.</p><p><strong>Conclusions: </strong>Both pasireotide formulations showed dose-proportional pharmacokinetics and 300-900 μg of SC pasireotide and 20-60 mg LAR pasireotide treatment showed favorable safety profiles and was well-tolerated when administered in male Taiwanese volunteers who are hyperendemic hepatitis B/C and CKD.</p>","PeriodicalId":11939,"journal":{"name":"European Journal of Drug Metabolism and Pharmacokinetics","volume":" ","pages":"665-674"},"PeriodicalIF":1.9,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41117325","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-11-01Epub Date: 2023-10-04DOI: 10.1007/s13318-023-00855-3
Joel S Owen, Russell J Rackley, Matthew A Hummel, Stefan Roepcke, Hannah Huang, Mark Liu, Tazeen A Idris, Sundara Moorthi Nainar Murugesan, Ashwani Marwah, Subramanian Loganathan, Gopinath Ranganna, Abhijit Barve, Cornelius F Waller, Mark A Socinski
Background and objectives: MYL-1402O is a bevacizumab (Avastin®) biosimilar. Pharmacokinetic and safety similarity of MYL-1402O and reference Avastin® authorized in the European Union (EU-Avastin®) and the US (US-Avastin®) was demonstrated in healthy subjects (phase I, NCT02469987). The key objectives of this study were to establish a population pharmacokinetic (PopPK) model on pooled data from the phase I and phase III clinical studies to assess pharmacokinetic linearity of MYL-1402O and Avastin® across dose ranges, to assess the pharmacokinetic similarity of MYL-1402O and Avastin® in patients with non-squamous non-small cell lung cancer (nsNSCLC), and to explore potential covariates to account for systematic sources of variability in bevacizumab exposure.
Methods: Efficacy and safety of MYL-1402O compared with EU-Avastin® was investigated in a multicenter, double-blind, randomized, parallel-group study in patients with stage IV nsNSCLC (phase III, NCT04633564). PopPK models were developed using a nonlinear mixed effects approach (NONMEM® 7.3.0).
Results: The pharmacokinetics of Avastin® and MYL-1402O were adequately described with a two-compartment linear model. Fourteen covariates were found to be statistically significant predictors of bevacizumab pharmacokinectics. The impact of each covariate on area under the concentration-time curve, half-life, and maximum plasma concentration was modest, and ranges were similar between the treatment groups, MYL-1402O and EU-Avastin®, in patients with nsNSCLC. The pharmacokinectics of bevacizumab appeared to be linear.
Conclusions: PopPK analysis revealed no significant differences between pharmacokinetics of MYL-1402O and Avastin® in patients with nsNSCLC. The developed PopPK model was considered robust, as it adequately described bevacizumab pharmacokinetics in healthy participants and nsNSCLC patients.
{"title":"Population Pharmacokinetics of MYL-1402O, a Proposed Biosimilar to Bevacizumab and Reference Product (Avastin<sup>®</sup>) in Patients with Non-squamous Non-small Cell Lung Cancer.","authors":"Joel S Owen, Russell J Rackley, Matthew A Hummel, Stefan Roepcke, Hannah Huang, Mark Liu, Tazeen A Idris, Sundara Moorthi Nainar Murugesan, Ashwani Marwah, Subramanian Loganathan, Gopinath Ranganna, Abhijit Barve, Cornelius F Waller, Mark A Socinski","doi":"10.1007/s13318-023-00855-3","DOIUrl":"10.1007/s13318-023-00855-3","url":null,"abstract":"<p><strong>Background and objectives: </strong>MYL-1402O is a bevacizumab (Avastin<sup>®</sup>) biosimilar. Pharmacokinetic and safety similarity of MYL-1402O and reference Avastin<sup>®</sup> authorized in the European Union (EU-Avastin<sup>®</sup>) and the US (US-Avastin<sup>®</sup>) was demonstrated in healthy subjects (phase I, NCT02469987). The key objectives of this study were to establish a population pharmacokinetic (PopPK) model on pooled data from the phase I and phase III clinical studies to assess pharmacokinetic linearity of MYL-1402O and Avastin<sup>®</sup> across dose ranges, to assess the pharmacokinetic similarity of MYL-1402O and Avastin<sup>®</sup> in patients with non-squamous non-small cell lung cancer (nsNSCLC), and to explore potential covariates to account for systematic sources of variability in bevacizumab exposure.</p><p><strong>Methods: </strong>Efficacy and safety of MYL-1402O compared with EU-Avastin<sup>®</sup> was investigated in a multicenter, double-blind, randomized, parallel-group study in patients with stage IV nsNSCLC (phase III, NCT04633564). PopPK models were developed using a nonlinear mixed effects approach (NONMEM<sup>®</sup> 7.3.0).</p><p><strong>Results: </strong>The pharmacokinetics of Avastin<sup>®</sup> and MYL-1402O were adequately described with a two-compartment linear model. Fourteen covariates were found to be statistically significant predictors of bevacizumab pharmacokinectics. The impact of each covariate on area under the concentration-time curve, half-life, and maximum plasma concentration was modest, and ranges were similar between the treatment groups, MYL-1402O and EU-Avastin<sup>®</sup>, in patients with nsNSCLC. The pharmacokinectics of bevacizumab appeared to be linear.</p><p><strong>Conclusions: </strong>PopPK analysis revealed no significant differences between pharmacokinetics of MYL-1402O and Avastin<sup>®</sup> in patients with nsNSCLC. The developed PopPK model was considered robust, as it adequately described bevacizumab pharmacokinetics in healthy participants and nsNSCLC patients.</p>","PeriodicalId":11939,"journal":{"name":"European Journal of Drug Metabolism and Pharmacokinetics","volume":" ","pages":"675-689"},"PeriodicalIF":1.9,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41144220","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background and objective: HSK21542, a synthetic short-chain polypeptide, is a selective peripheral kappa opioid receptor (KOR) agonist. In this single-centre, non-randomized, open-label study, the pharmacokinetics, mass balance, metabolism and excretion of HSK21542 were investigated.
Methods: A single intravenous dose of 2 μg/0.212 μCi/kg [14C]HSK21542 was administered to six healthy male subjects. Samples of blood, urine and faeces were collected for quantitative determination of total radioactivity and unchanged HSK21542, and identification of metabolites.
Results: The mean total recovery was 81.89% of the radiolabelled dose over 240 h post-dose, with 35.60% and 46.30% excreted in faeces and urine, respectively. The mean maximum concentration (Cmax), the half-life (t1/2) and the area under the concentration-time curve (AUC0-t) of total radioactivity (TRA) in plasma were 20.4 ±4.16 ng Eq./g, 1.93 ± 0.322 h and 21.8 ± 2.93 h·ng Eq./g, respectively, while the Cmax, t1/2 and the AUC0-t of unchanged HSK21542 were 18.3 ± 3.36 ng/mL, 1.66 ± 0.185 h and 18.4 ± 2.24 h·ng/mL, respectively. The blood-to-plasma ratios of TRA at several times ranged from 0.46 to 0.54. [14C]HSK21542 was detected as the main circulating substance in plasma, accounting for 92.17% of the AUC of TRA. The unchanged parent compound was the only major radioactive chemical in urine (100.00% of TRA) and faeces (93.53% of TRA). Metabolites were very minor components.
Conclusions: HSK21542 was barely metabolized in vivo and mainly excreted with unchanged HSK21542 as its main circulating component in plasma. It was speculated that renal excretion was the principal excretion pathway, and faecal excretion was the secondary pathway.
Clinical trial registration number: NCT05835934.
背景与目的:HSK21542是一种合成的短链多肽,是一种选择性外周κ阿片受体激动剂。在这项单中心、非随机、开放标签研究中,研究了HSK21542的药代动力学、质量平衡、代谢和排泄。方法:6名健康男性受试者单次静脉注射2μg/0.212μCi/kg[14C]HSK21542。采集血液、尿液和粪便样本,用于定量测定总放射性和不变的HSK21542,并鉴定代谢物。结果:在给药后240小时内,平均总回收率为放射性标记剂量的81.89%,其中35.60%和46.30%分别通过粪便和尿液排出。血浆中总放射性(TRA)的平均最大浓度(Cmax)、半衰期(t1/2)和浓度-时间曲线下面积(AUC0-t)分别为20.4±4.16 ng Eq./g、1.93±0.322 h和21.8±2.93 h·ng Eq./g,而不变的HSK21542的Cmax、t1/2和AUC0-t分别为18.3±3.36 ng/mL、1.66±0.185 h和18.4±2.24 h·ng/mL,分别地TRA的血浆比在几个时间范围内为0.46至0.54。[14C]HSK21542为血浆中主要循环物质,占TRA AUC的92.17%。不变的母体化合物是尿液(占TRA的100.00%)和粪便(占TRA的93.53%)中唯一的主要放射性化学物质。代谢物是非常次要的成分。结论:HSK21542在体内几乎没有代谢,主要以血浆中HSK21542-为主要循环成分而排泄。推测肾脏排泄是主要排泄途径,粪便排泄是次要排泄途径。临床试验注册号:NCT05835934。
{"title":"Pharmacokinetics, Mass Balance and Metabolism of [<sup>14</sup>C]HSK21542, a Novel Kappa Opioid Receptor Agonist, in Humans.","authors":"Jin-Jie Yuan, Yi-Cong Bian, Sheng Ma, Wei Chen, Feng-Yi Zhang, Hua Zhang, Li-Yan Miao","doi":"10.1007/s13318-023-00858-0","DOIUrl":"10.1007/s13318-023-00858-0","url":null,"abstract":"<p><strong>Background and objective: </strong>HSK21542, a synthetic short-chain polypeptide, is a selective peripheral kappa opioid receptor (KOR) agonist. In this single-centre, non-randomized, open-label study, the pharmacokinetics, mass balance, metabolism and excretion of HSK21542 were investigated.</p><p><strong>Methods: </strong>A single intravenous dose of 2 μg/0.212 μCi/kg [<sup>14</sup>C]HSK21542 was administered to six healthy male subjects. Samples of blood, urine and faeces were collected for quantitative determination of total radioactivity and unchanged HSK21542, and identification of metabolites.</p><p><strong>Results: </strong>The mean total recovery was 81.89% of the radiolabelled dose over 240 h post-dose, with 35.60% and 46.30% excreted in faeces and urine, respectively. The mean maximum concentration (C<sub>max</sub>), the half-life (t<sub>1/2</sub>) and the area under the concentration-time curve (AUC<sub>0-t</sub>) of total radioactivity (TRA) in plasma were 20.4 ±4.16 ng Eq./g, 1.93 ± 0.322 h and 21.8 ± 2.93 h·ng Eq./g, respectively, while the C<sub>max</sub>, t<sub>1/2</sub> and the AUC<sub>0-t</sub> of unchanged HSK21542 were 18.3 ± 3.36 ng/mL, 1.66 ± 0.185 h and 18.4 ± 2.24 h·ng/mL, respectively. The blood-to-plasma ratios of TRA at several times ranged from 0.46 to 0.54. [<sup>14</sup>C]HSK21542 was detected as the main circulating substance in plasma, accounting for 92.17% of the AUC of TRA. The unchanged parent compound was the only major radioactive chemical in urine (100.00% of TRA) and faeces (93.53% of TRA). Metabolites were very minor components.</p><p><strong>Conclusions: </strong>HSK21542 was barely metabolized in vivo and mainly excreted with unchanged HSK21542 as its main circulating component in plasma. It was speculated that renal excretion was the principal excretion pathway, and faecal excretion was the secondary pathway.</p><p><strong>Clinical trial registration number: </strong>NCT05835934.</p>","PeriodicalId":11939,"journal":{"name":"European Journal of Drug Metabolism and Pharmacokinetics","volume":" ","pages":"723-731"},"PeriodicalIF":1.9,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41195599","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2023-11-01Epub Date: 2023-10-13DOI: 10.1007/s13318-023-00860-6
Shyamala Mudavath, Dongamanti Ashok
Background and objectives: Ripretinib was developed to target a whole range of KIT proto-oncogene mutations and platelet-derived growth factor receptor A (PDGFR-A) kinases found in certain cancers and myeloproliferative neoplasms, particularly gastrointestinal stromal tumours (GISTs). This study investigated the effect of verapamil, a potential inhibitor of P-glycoprotein-1 (P-gp1) and cytochrome P450 3A4 (CYP3A4), on the pharmacokinetics of ripretinib in rats when administered orally together. This study also assessed the metabolic stability and in vitro cellular absorption of ripretinib in the presence of verapamil.
Methods: A novel sensitive time-saving liquid chromatography tandem mass spectometry (LC-MS/MS) technique for determining ripretinib in rat plasma was developed and validated. A Zorbax SB C18 column was used for the separation and analysis of ripretinib with a mobile phase consisting of 50:50 (%v/v) acetonitrile and 10 mM ammonium formate buffer at a flow rate of 0.4 mL/min. Imatinib was used as an internal standard (IS) in the method. The pharmacokinetic characteristics of ripretinib were evaluated in Wistar rats by successfully administering an oral dosage of 5 mg/kg body weight of ripretinib in the presence of verapamil (10 mg/kg body weight). Subsequently, rat liver microsomes were used to assess the effect of verapamil on ripretinib metabolic stability, and absorption was tested using a Caco-2 cell transwell model.
Results: Ripretinib and IS were identified using multiple reaction monitoring (MRM) modes by mass spectrometry and showed ion transitions of 510.09→94.06 m/z and 494.26→ 394.16 m/z, respectively. The high-performance liquid chromatography (HPLC) method successfully eluted ripretinib and IS at retention times of 0.91 and 0.68 min, respectively, and the method was validated for all parameters and met the criteria for acceptance. Co-administration of verapamil increased the maximum concentration (Cmax) of ripretinib from 437 ± 84 ng/mL to 492 ± 50 ng/mL (12%), and the area under the concentration-time curve from 0 to the last sampling time t (AUC0-t) increased by approximately 40.6%. Verapamil significantly reduced the basolateral-to-apical transfer of ripretinib through Caco-2 cells. Findings also showed that verapamil increased the metabolic stability of ripretinib.
Conclusion: The study results indicate that the co-administration of ripretinib with CYP3A4 and/or P-gp1 inhibitors is associated with significant drug-drug interactions that affect the pharmacokinetics of ripretinib. Further research in human subjects is suggested to confirm dosage adjustment and therapeutic drug monitoring of ripretinib when administered along with P-gp1/CYP3A4 inhibitors ensuring patient safety and optimizing the therapeutic benefits of ripretinib.
{"title":"Effect of Verapamil, a P-glycoprotein-1 and Cytochrome P450 3A4 Inhibitor, on Pharmacokinetics and Metabolic Stability of Ripretinib: A Drug-Drug Interaction Study in Rats.","authors":"Shyamala Mudavath, Dongamanti Ashok","doi":"10.1007/s13318-023-00860-6","DOIUrl":"10.1007/s13318-023-00860-6","url":null,"abstract":"<p><strong>Background and objectives: </strong>Ripretinib was developed to target a whole range of KIT proto-oncogene mutations and platelet-derived growth factor receptor A (PDGFR-A) kinases found in certain cancers and myeloproliferative neoplasms, particularly gastrointestinal stromal tumours (GISTs). This study investigated the effect of verapamil, a potential inhibitor of P-glycoprotein-1 (P-gp1) and cytochrome P450 3A4 (CYP3A4), on the pharmacokinetics of ripretinib in rats when administered orally together. This study also assessed the metabolic stability and in vitro cellular absorption of ripretinib in the presence of verapamil.</p><p><strong>Methods: </strong>A novel sensitive time-saving liquid chromatography tandem mass spectometry (LC-MS/MS) technique for determining ripretinib in rat plasma was developed and validated. A Zorbax SB C18 column was used for the separation and analysis of ripretinib with a mobile phase consisting of 50:50 (%v/v) acetonitrile and 10 mM ammonium formate buffer at a flow rate of 0.4 mL/min. Imatinib was used as an internal standard (IS) in the method. The pharmacokinetic characteristics of ripretinib were evaluated in Wistar rats by successfully administering an oral dosage of 5 mg/kg body weight of ripretinib in the presence of verapamil (10 mg/kg body weight). Subsequently, rat liver microsomes were used to assess the effect of verapamil on ripretinib metabolic stability, and absorption was tested using a Caco-2 cell transwell model.</p><p><strong>Results: </strong>Ripretinib and IS were identified using multiple reaction monitoring (MRM) modes by mass spectrometry and showed ion transitions of 510.09→94.06 m/z and 494.26→ 394.16 m/z, respectively. The high-performance liquid chromatography (HPLC) method successfully eluted ripretinib and IS at retention times of 0.91 and 0.68 min, respectively, and the method was validated for all parameters and met the criteria for acceptance. Co-administration of verapamil increased the maximum concentration (C<sub>max</sub>) of ripretinib from 437 ± 84 ng/mL to 492 ± 50 ng/mL (12%), and the area under the concentration-time curve from 0 to the last sampling time t (AUC<sub>0-t</sub>) increased by approximately 40.6%. Verapamil significantly reduced the basolateral-to-apical transfer of ripretinib through Caco-2 cells. Findings also showed that verapamil increased the metabolic stability of ripretinib.</p><p><strong>Conclusion: </strong>The study results indicate that the co-administration of ripretinib with CYP3A4 and/or P-gp1 inhibitors is associated with significant drug-drug interactions that affect the pharmacokinetics of ripretinib. Further research in human subjects is suggested to confirm dosage adjustment and therapeutic drug monitoring of ripretinib when administered along with P-gp1/CYP3A4 inhibitors ensuring patient safety and optimizing the therapeutic benefits of ripretinib.</p>","PeriodicalId":11939,"journal":{"name":"European Journal of Drug Metabolism and Pharmacokinetics","volume":" ","pages":"733-746"},"PeriodicalIF":1.9,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41195598","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}