1
|
Nishioka K, Ishimoto T, Sato M, Yasuda R, Nakamura Y, Watanabe H, Suzuki T, Araragi Y, Kato Y, Yoshida KI, Murayama N. Optimizing oral 3-hydroxybutyrate dosage using pharmacokinetic model to improve cognitive function and mood in healthy subjects. Front Nutr 2025; 11:1470331. [PMID: 39867559 PMCID: PMC11758625 DOI: 10.3389/fnut.2024.1470331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 12/11/2024] [Indexed: 01/28/2025] Open
Abstract
Introduction The brain uses ketones, mainly 3-hydroxybutyrate (3-HB), as an alternative energy source. Therefore, oral intake of 3-HB may help maintain brain health. Previous studies indicated that achieving a maximum concentration (Cmax) of 3-HB in plasma at 0.28 mM could initiate ketone metabolism in the brain; we hypothesized that attaining this Cmax would improve brain health. Methods We aimed to demonstrate the efficacy of an optimized single oral dose of 3-HB on cognitive function and mood through two clinical studies: a pharmacokinetic study and an efficacy study. In the pharmacokinetic study, healthy subjects were ingested 2 and 4 g of 3-HB to construct a compartment model to predict the minimum oral dose of 3-HB needed to achieve the target Cmax. In the efficacy study, a randomized, double-blinded, and placebo-controlled crossover trial, the effects of 3-HB at the predicted doses on cognitive function and mood in healthy subjects were assessed by a serial arithmetic test (SAT), the cognitrax, the profile of mood states 2nd edition (POMS2), and fatigue visual analog scale (VAS). Results In the pharmacokinetic study, a one-compartment model that includes saturable and non-saturable absorption pathways, constant biosynthesis, and the linear elimination of 3-HB after oral administration were constructed. The model principally reflected the observed serum 3-HB concentrations profiles and predicted a minimum dose of 3.5 g needed to achieve the target Cmax. In the efficacy study, although no significant difference was observed in any cognitive domains assessed by the Cognitrax, total responses and correct answers in the SAT were significantly improved in the active group receiving 3.5 g of 3-HB compared to the placebo group. Regarding the POMS2, confusion-bewilderment, fatigue-inertia, vigor-activity, and total mood disturbance scales were significantly improved in the active group compared to the placebo group. Additionally, fatigue VAS were also significantly improved in the active group compared to the placebo group. Discussion We successfully established a one-compartment model for oral 3-HB intake and demonstrated partial efficacy on cognitive function and broad efficacy on mood in healthy subjects with a single oral dose of 3.5 g of 3-HB optimized by the model. Clinical trial registration https://www.umin.ac.jp/ctr/index-j.htm, identifier [UMIN000042095, UMIN000046666].
Collapse
Affiliation(s)
- Kentaro Nishioka
- Research Institute, Suntory Global Innovation Center Ltd., Kyoto, Japan
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
- Department of Science, Technology and Innovation, Kobe University, Kobe, Japan
| | - Takahiro Ishimoto
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Mariko Sato
- Research Institute, Suntory Global Innovation Center Ltd., Kyoto, Japan
| | - Ruki Yasuda
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Yumi Nakamura
- Research Institute, Suntory Global Innovation Center Ltd., Kyoto, Japan
| | - Hiroshi Watanabe
- Research Institute, Suntory Global Innovation Center Ltd., Kyoto, Japan
| | - Toshihide Suzuki
- Research Institute, Suntory Global Innovation Center Ltd., Kyoto, Japan
| | - Yudai Araragi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Ken-ichi Yoshida
- Department of Science, Technology and Innovation, Kobe University, Kobe, Japan
| | - Norihito Murayama
- Research Institute, Suntory Global Innovation Center Ltd., Kyoto, Japan
| |
Collapse
|
2
|
Mizuno S, Takabayashi M, Makihara H, Ogai K, Tsukui K, Ito Y, Kawakami T, Hara Y, Fujita A, Tokudome Y, Akase T, Kato Y, Shimada T, Sai Y. Effect of changes in skin properties due to diabetes mellitus on the titration period of transdermal fentanyl: single-center retrospective study and diabetic animal model study. J Pharm Health Care Sci 2024; 10:80. [PMID: 39696707 DOI: 10.1186/s40780-024-00402-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND In the dose titration of transdermal fentanyl to prevent unrelieved pain, it is important to consider not only dose adjustment, but also the titration period, which is influenced by the time required to reach the steady state. Many patients with cancer pain experience comorbidities that might affect the skin properties and influence transdermal absorption. We hypothesized that skin changes due to diabetes mellitus (DM) would affect the titration period of transdermal fentanyl. We conducted a retrospective study and diabetic animal model study to test this hypothesis. METHODS In the retrospective study, the titration period was defined in terms of "dose change" and "number of rescue opioids" in patients initiated on transdermal fentanyl. Multiple logistic regression analysis was performed to analyze the relation between the titration period and comorbidities, including DM. In the diabetic animal model study, intercellular lipids of stratum corneum (SC) were analyzed in Goto-Kakizaki (GK) rats, a model of DM, and the pharmacokinetics of intravenously or transdermally administered fentanyl was examined. RESULTS In the retrospective study, the titration period ranged from 5 to 39 days (n = 387), and the patients taking a longer period (6 days or more) was significantly related to in patients with unspecified DM: AOR (95% confidence interval), 0.438 (0.217-0.884). In the diabetic animal model study, the ceramides (CERs) content in the SC was decreased by approximately 30% in GK rats compared to Wistar rats. The absorption rate constant (ka) of fentanyl administered transdermally was increased approximately 1.4-fold in GK rats, though there was no difference in transdermal bioavailability (F) or systemic clearance (CLtot). CONCLUSION Our results suggest that the steady state of transdermally administered fentanyl is reached sooner in cancer patients with DM as a comorbidity. Earlier pain assessment and dose adjustment may be possible in these patients.
Collapse
Affiliation(s)
- Satoshi Mizuno
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Makiko Takabayashi
- Department of Hospital Pharmacy, University Hospital, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Hiroko Makihara
- Department of Biochemistry, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
- Department of Biological Science and Nursing, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Kazuhiro Ogai
- Department of Bio-Engineering Nursing, Graduate School of Nursing, Ishikawa Prefectural Nursing University, Kahoku, Japan
| | - Kei Tsukui
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| | - Yuriko Ito
- Department of Hospital Pharmacy, University Hospital, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Takahiro Kawakami
- Department of Hospital Pharmacy, University Hospital, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Yusuke Hara
- Department of Hospital Pharmacy, University Hospital, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Arimi Fujita
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
- Department of Hospital Pharmacy, University Hospital, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Yoshihiro Tokudome
- Laboratory of Cosmetic Sciences, Institute of Ocean Energy, Saga University, Saga, Japan
| | - Tomoko Akase
- Department of Biological Science and Nursing, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Yukio Kato
- Department of Molecular Pharmacotherapeutics, Faculty of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Tsutomu Shimada
- Department of Clinical Pharmacy and Healthcare Science, Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Science, Kanazawa University, Kanazawa, Japan
| | - Yoshimichi Sai
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan.
- Department of Hospital Pharmacy, University Hospital, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan.
- AI Hospital/Macro Signal Dynamics Research and Development Center, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
3
|
Mizuno S, Gake S, Takabayashi M, Ito Y, Sanada H, Sugimoto N, Maeda A, Tamamura T, Sawamoto K, Hara Y, Ohi Y, Tsuji C, Shiomoto Y, Kato Y, Fujita A, Shimada T, Miyamoto KI, Sai Y. Effect of obesity on pharmacokinetics of transdermal fentanyl: Single-center retrospective study and animal study. Drug Metab Pharmacokinet 2024; 60:101035. [PMID: 39693856 DOI: 10.1016/j.dmpk.2024.101035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/19/2024] [Accepted: 11/03/2024] [Indexed: 12/20/2024]
Abstract
A retrospective study and an animal study were conducted to investigate factors affecting the transdermal fentanyl dose to achieve adequate pain relief in patients switched from other opioids. In the retrospective study, patient factors were included as gender, age, body mass index (BMI), and serum albumin concentration. In obese (BMI ≥25) patients, the post-titration dose of transdermal fentanyl was significantly lower than in normal (BMI 18.5-25) patients despite the initial dose was the same. To support this unexpected finding, fentanyl was administered intravenously and transdermally to Zucker (fa/fa) rats as an obese model and Zucker (+/+) rats as a control. No difference in systemic clearance (CLtot) after intravenous administration was observed between the two groups. However, transdermal bioavailability (F) and fentanyl release ratio from the formulation (Fa) were significantly increased in Zucker (fa/fa) rats compared to Zucker (+/+) rats. Skin availability (Fskin), calculated as F divided by Fa, was also significantly increased. These results indicated that obesity should be considered as a factor in the titration of transdermal fentanyl dose.
Collapse
Affiliation(s)
- Satoshi Mizuno
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Shintaro Gake
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Makiko Takabayashi
- Department of Hospital Pharmacy, University Hospital, Kanazawa University, Kanazawa, Japan
| | - Yuriko Ito
- Department of Hospital Pharmacy, University Hospital, Kanazawa University, Kanazawa, Japan
| | - Hiroko Sanada
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Natsumi Sugimoto
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Akari Maeda
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Takuto Tamamura
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kazuki Sawamoto
- Department of Hospital Pharmacy, University Hospital, Kanazawa University, Kanazawa, Japan
| | - Yusuke Hara
- Department of Hospital Pharmacy, University Hospital, Kanazawa University, Kanazawa, Japan
| | - Yoshiko Ohi
- Department of Hospital Pharmacy, University Hospital, Kanazawa University, Kanazawa, Japan
| | - Chiaki Tsuji
- Department of Hospital Pharmacy, University Hospital, Kanazawa University, Kanazawa, Japan
| | - Yukiko Shiomoto
- Department of Hospital Pharmacy, University Hospital, Kanazawa University, Kanazawa, Japan
| | - Yukio Kato
- Department of Molecular Pharmacotherapeutics, Faculty of Pharmacy, Kanazawa University, Kanazawa, Japan
| | - Arimi Fujita
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan; Department of Hospital Pharmacy, University Hospital, Kanazawa University, Kanazawa, Japan
| | - Tsutomu Shimada
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan; Department of Hospital Pharmacy, University Hospital, Kanazawa University, Kanazawa, Japan; Department of Clinical Pharmacy and Healthcare Science, Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Science, Kanazawa University, Kanazawa, Japan
| | - Ken-Ichi Miyamoto
- Department of Hospital Pharmacy, University Hospital, Kanazawa University, Kanazawa, Japan
| | - Yoshimichi Sai
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan; Department of Hospital Pharmacy, University Hospital, Kanazawa University, Kanazawa, Japan; AI Hospital/Macro Signal Dynamics Research and Development Center, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
4
|
Tsuchitani T, Tomaru A, Aoki Y, Ishiguro N, Tsuda Y, Sugiyama Y. Elucidating nonlinear pharmacokinetics of telmisartan: Integration of target-mediated drug disposition and OATP1B3-mediated hepatic uptake in a physiologically based model. CPT Pharmacometrics Syst Pharmacol 2024; 13:1224-1237. [PMID: 38745377 PMCID: PMC11247111 DOI: 10.1002/psp4.13154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/25/2024] [Accepted: 04/16/2024] [Indexed: 05/16/2024] Open
Abstract
Telmisartan, a selective inhibitor of angiotensin II receptor type 1 (AT1), demonstrates nonlinear pharmacokinetics (PK) when orally administered in ascending doses to healthy volunteers, but the underlying mechanisms remain unclear. This study presents a physiologically based pharmacokinetic model integrated with target-mediated drug disposition (TMDD-PBPK model) to explore the mechanism of its nonlinear PK. We employed the Cluster-Gauss Newton method for top-down analysis, estimating the in vivo Km,OATP1B3 (Michaelis-Menten constant for telmisartan hepatic uptake via Organic Anion Transporting Polypeptide 1B3) to be 2.0-5.7 nM. This range is significantly lower than the reported in vitro value of 810 nM, obtained in 0.3% human serum albumin (HSA) conditions. Further validation was achieved through in vitro assessment in plated human hepatocytes with 4.5% HSA, showing a Km of 4.5 nM. These results underscore the importance of albumin-mediated uptake effect for the hepatic uptake of telmisartan. Our TMDD-PBPK model, developed through a "middle-out" approach, underwent sensitivity analysis to identify key factors in the nonlinear PK of telmisartan. We found that the nonlinearity in the area under the concentration-time curve (AUC) and/or maximum concentration (Cmax) of telmisartan is sensitive to Km,OATP1B3 across all dosages. Additionally, the dissociation constant (Kd) for telmisartan binding to the AT1 receptor, along with its receptor abundance, notably influences PK at lower doses (below 20 mg). In conclusion, the nonlinear PK of telmisartan appears primarily driven by hepatic uptake saturation across all dose ranges and by AT1-receptor binding saturation, notably at lower doses.
Collapse
Affiliation(s)
- Toshiaki Tsuchitani
- iHuman InstituteShanghaiTech UniversityShanghaiChina
- Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, School of PharmacyJosai International University (JIU)TokyoJapan
| | - Atsuko Tomaru
- Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, School of PharmacyJosai International University (JIU)TokyoJapan
| | - Yasunori Aoki
- Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, School of PharmacyJosai International University (JIU)TokyoJapan
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM)BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Naoki Ishiguro
- Pharmacokinetics and Non‐Clinical Safety DepartmentNippon Boehringer Ingelheim Co., Ltd.KobeHyogoJapan
| | - Yasuhiro Tsuda
- Clinical Pharmacology DepartmentNippon Boehringer Ingelheim Co., Ltd.KobeHyogoJapan
| | - Yuichi Sugiyama
- iHuman InstituteShanghaiTech UniversityShanghaiChina
- Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, School of PharmacyJosai International University (JIU)TokyoJapan
| |
Collapse
|
5
|
Toshimoto K. Beyond the basics: A deep dive into parameter estimation for advanced PBPK and QSP models. Drug Metab Pharmacokinet 2024; 56:101011. [PMID: 38833901 DOI: 10.1016/j.dmpk.2024.101011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/26/2024] [Accepted: 03/14/2024] [Indexed: 06/06/2024]
Abstract
Physiologically-based pharmacokinetic (PBPK) models and quantitative systems pharmacology (QSP) models have contributed to drug development strategies. The parameters of these models are commonly estimated by capturing observed values using the nonlinear least-squares method. Software packages for PBPK and QSP modeling provide a range of parameter estimation algorithms. To choose the most appropriate method, modelers need to understand the basic concept of each approach. This review provides a general introduction to the key points of parameter estimation with a focus on the PBPK and QSP models, and the respective parameter estimation algorithms. The latter part assesses the performance of five parameter estimation algorithms - the quasi-Newton method, Nelder-Mead method, genetic algorithm, particle swarm optimization, and Cluster Gauss-Newton method - using three examples of PBPK and QSP modeling. The assessment revealed that some parameter estimation results were significantly influenced by the initial values. Moreover, the choice of algorithms demonstrating good estimation results heavily depends on factors such as model structure and the parameters to be estimated. To obtain credible parameter estimation results, it is advisable to conduct multiple rounds of parameter estimation under different conditions, employing various estimation algorithms.
Collapse
Affiliation(s)
- Kota Toshimoto
- Systems Pharmacology, Non-Clinical Biomedical Science, Applied Research & Operations, Astellas Pharma Inc., Ibaraki, Japan.
| |
Collapse
|
6
|
Okubo K, Kudo T, Yoshihara S, Nakabayashi Y, Nakauchi K, Tanaka A, Saito M, Tsujisawa A, Goda H, Yamagishi Y, Otake C, Makino K, Takahashi H, Ito K. Physiologically based pharmacokinetic model analysis of the inhibitory effect of vonoprazan on the metabolic activation of proguanil. Drug Metab Pharmacokinet 2024; 54:100537. [PMID: 38086197 DOI: 10.1016/j.dmpk.2023.100537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/15/2023] [Accepted: 10/30/2023] [Indexed: 02/06/2024]
Abstract
We previously reported that repeated oral administration of vonoprazan (VPZ) followed by oral administration of proguanil (PG) in healthy adults increased blood concentration of PG and decreased blood concentration of its metabolite cycloguanil (CG) compared with administration of PG alone. In this study, we investigated whether this interaction can be quantitatively explained by VPZ inhibition of PG metabolism. In an in vitro study using human liver microsomes, VPZ inhibited CG formation from PG in a concentration-dependent manner, and the inhibition was enhanced depending on preincubation time. Then, a physiologically based pharmacokinetic (PBPK) model analysis was performed incorporating the obtained inhibition parameters. By fitting the blood concentration profiles of VPZ and PG/CG after VPZ and PG were orally administered alone to our PBPK model, parameters were obtained which can reproduce their concentration profiles. In contrast, when the VPZ inhibition parameters for CG formation from the in vitro study were incorporated, the predicted blood PG and CG concentrations were unchanged; the apparent dissociation constant had to be set to about 1/23 of the obtained in vitro value to reproduce the observed interaction. Further comprehensive evaluation is required, including the possibility that mechanisms other than metabolic inhibition may be involved.
Collapse
Affiliation(s)
- Kenjiro Okubo
- Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585, Japan
| | - Toshiyuki Kudo
- Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585, Japan; Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585, Japan.
| | - Sae Yoshihara
- Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585, Japan
| | - Yu Nakabayashi
- Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585, Japan
| | - Kana Nakauchi
- Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585, Japan
| | - Akimi Tanaka
- Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585, Japan
| | - Moe Saito
- Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585, Japan
| | - Ayumi Tsujisawa
- Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585, Japan
| | - Hitomi Goda
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585, Japan
| | - Yoshiaki Yamagishi
- Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585, Japan; Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585, Japan.
| | - Chinatsu Otake
- Department of Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda-shi, Chiba, 278-8510, Japan
| | - Kosho Makino
- Department of Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda-shi, Chiba, 278-8510, Japan.
| | - Hideyo Takahashi
- Department of Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda-shi, Chiba, 278-8510, Japan.
| | - Kiyomi Ito
- Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585, Japan; Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585, Japan.
| |
Collapse
|
7
|
Suzuki S, Inoue K, Tamai I, Shirasaka Y. Quantitative Analysis of Gastrointestinal Water Dynamics by Means of a Physiologically Based Fluid Kinetic Model. AAPS J 2023; 25:42. [PMID: 37081157 DOI: 10.1208/s12248-023-00809-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/28/2023] [Indexed: 04/22/2023] Open
Abstract
Since the processes of dissolution and membrane permeation are affected by the water content in the gastrointestinal (GI) tract, the water dynamics in the GI tract is expected to have a significant impact on the absorption of orally administered drugs. Here, we aimed to develop a physiologically based fluid kinetic (PBFK) model using GI water kinetic parameters obtained from in situ closed-loop studies in rats in order to quantitatively predict GI water dynamics. By incorporating the experimentally measured site-specific parameters of GI water absorption and secretion into a GI compartment model, we developed a bottom-up PBFK model that successfully simulates the reported GI fluid dynamics in rats and humans observed using positron emission tomography and magnetic resonance imaging, respectively. The simulations indicate that the water volume in both the stomach and duodenum is transiently increased by water ingestion, while that in the intestine below the jejunum is unchanged and remains in a steady state in both rats and humans. Furthermore, sensitivity analysis of the effect of ingested water volume on the volume-time profiles of water in the GI tract indicated that the impact of ingested water is limited to the proximal part of the GI tract. Simulations indicated that changes in water kinetic parameters may alter the impact of the ingested water on GI fluid dynamics, especially in the proximal part. Incorporating this PBFK model into a physiologically based pharmacokinetic (PBPK) absorption model has the potential to predict oral drug absorption in a variety of GI water environments.
Collapse
Affiliation(s)
- Satoru Suzuki
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, 920-1192, Japan
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Tokyo, 192-0392, Japan
| | - Katsuhisa Inoue
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Tokyo, 192-0392, Japan
| | - Ikumi Tamai
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, 920-1192, Japan
| | - Yoshiyuki Shirasaka
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, 920-1192, Japan.
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Tokyo, 192-0392, Japan.
| |
Collapse
|
8
|
Yoshitomo A, Asano S, Hozuki S, Tamemoto Y, Shibata Y, Hashimoto N, Takahashi K, Sasaki Y, Ozawa N, Kageyama M, Iijima T, Kazuki Y, Sato H, Hisaka A. Significance of Basal Membrane Permeability of Epithelial Cells in Predicting Intestinal Drug Absorption. Drug Metab Dispos 2023; 51:318-328. [PMID: 36810197 DOI: 10.1124/dmd.122.000907] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Drug absorption from the gastrointestinal tract is often restricted by efflux transport by P-glycoprotein (P-gp) and metabolism by CYP3A4. Both localize in the epithelial cells, and thus, their activities are directly affected by the intracellular drug concentration, which should be regulated by the ratio of permeability between apical (A) and basal (B) membranes. In this study, using Caco-2 cells with forced expression of CYP3A4, we assessed the transcellular permeation of A-to-B and B-to-A directions and the efflux from the preloaded cells to both sides of 12 representative P-gp or CYP3A4 substrate drugs and obtained the parameters for permeabilities, transport, metabolism, and unbound fraction in the enterocytes (fent) using simultaneous and dynamic model analysis. The membrane permeability ratios for B to A (RBA) and fent varied by 8.8-fold and by more than 3000-fold, respectively, among the drugs. The RBA values for digoxin, repaglinide, fexofenadine, and atorvastatin were greater than 1.0 (3.44, 2.39, 2.27, and 1.90, respectively) in the presence of a P-gp inhibitor, thus suggesting the potential involvement of transporters in the B membrane. The Michaelis constant for quinidine for P-gp transport was 0.077 µM for the intracellular unbound concentration. These parameters were used to predict overall intestinal availability (FAFG) by applying an intestinal pharmacokinetic model, advanced translocation model (ATOM), in which permeability of A and B membranes accounted separately. The model predicted changes in the absorption location for P-gp substrates according to its inhibition, and FAFG values of 10 of 12 drugs, including quinidine at varying doses, were explained appropriately. SIGNIFICANCE STATEMENT: Pharmacokinetics has improved predictability by identifying the molecular entities of metabolism and transport and by using mathematical models to appropriately describe drug concentrations at the locations where they act. However, analyses of intestinal absorption so far have not been able to accurately consider the concentrations in the epithelial cells where P-glycoprotein and CYP3A4 exert effects. In this study, the limitation was removed by measuring the apical and basal membrane permeability separately and then analyzing these values using new appropriate models.
Collapse
Affiliation(s)
- Aoi Yoshitomo
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.Y., S.A., S.H., Y.T., N.H., K.T., H.S., A.H.); Toxicology & DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A., Y.Sa., N.O., M.K., T.I.); Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (Y.Sh.); and Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Satoshi Asano
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.Y., S.A., S.H., Y.T., N.H., K.T., H.S., A.H.); Toxicology & DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A., Y.Sa., N.O., M.K., T.I.); Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (Y.Sh.); and Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Shizuka Hozuki
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.Y., S.A., S.H., Y.T., N.H., K.T., H.S., A.H.); Toxicology & DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A., Y.Sa., N.O., M.K., T.I.); Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (Y.Sh.); and Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Yuta Tamemoto
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.Y., S.A., S.H., Y.T., N.H., K.T., H.S., A.H.); Toxicology & DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A., Y.Sa., N.O., M.K., T.I.); Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (Y.Sh.); and Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Yukihiro Shibata
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.Y., S.A., S.H., Y.T., N.H., K.T., H.S., A.H.); Toxicology & DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A., Y.Sa., N.O., M.K., T.I.); Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (Y.Sh.); and Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Natsumi Hashimoto
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.Y., S.A., S.H., Y.T., N.H., K.T., H.S., A.H.); Toxicology & DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A., Y.Sa., N.O., M.K., T.I.); Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (Y.Sh.); and Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Keita Takahashi
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.Y., S.A., S.H., Y.T., N.H., K.T., H.S., A.H.); Toxicology & DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A., Y.Sa., N.O., M.K., T.I.); Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (Y.Sh.); and Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Yoko Sasaki
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.Y., S.A., S.H., Y.T., N.H., K.T., H.S., A.H.); Toxicology & DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A., Y.Sa., N.O., M.K., T.I.); Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (Y.Sh.); and Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Naoka Ozawa
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.Y., S.A., S.H., Y.T., N.H., K.T., H.S., A.H.); Toxicology & DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A., Y.Sa., N.O., M.K., T.I.); Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (Y.Sh.); and Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Michiharu Kageyama
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.Y., S.A., S.H., Y.T., N.H., K.T., H.S., A.H.); Toxicology & DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A., Y.Sa., N.O., M.K., T.I.); Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (Y.Sh.); and Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Takeshi Iijima
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.Y., S.A., S.H., Y.T., N.H., K.T., H.S., A.H.); Toxicology & DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A., Y.Sa., N.O., M.K., T.I.); Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (Y.Sh.); and Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Yasuhiro Kazuki
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.Y., S.A., S.H., Y.T., N.H., K.T., H.S., A.H.); Toxicology & DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A., Y.Sa., N.O., M.K., T.I.); Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (Y.Sh.); and Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Hiromi Sato
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.Y., S.A., S.H., Y.T., N.H., K.T., H.S., A.H.); Toxicology & DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A., Y.Sa., N.O., M.K., T.I.); Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (Y.Sh.); and Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Akihiro Hisaka
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.Y., S.A., S.H., Y.T., N.H., K.T., H.S., A.H.); Toxicology & DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A., Y.Sa., N.O., M.K., T.I.); Department of Regulatory Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (Y.Sh.); and Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| |
Collapse
|
9
|
Matsumoto T, Masuo Y, Tanaka A, Kimura T, Ioroi T, Yamakawa T, Kitahara H, Kato Y. A physiologically based pharmacokinetic and pharmacodynamic model for disposition of FF-10832. Int J Pharm 2022; 627:122250. [DOI: 10.1016/j.ijpharm.2022.122250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/29/2022] [Accepted: 09/24/2022] [Indexed: 10/31/2022]
|
10
|
Asaumi R, Nunoya K, Yamaura Y, Taskar KS, Sugiyama Y. Robust physiologically based pharmacokinetic model of rifampicin for predicting
drug–drug
interactions via P‐glycoprotein induction and inhibition in the intestine, liver, and kidney. CPT Pharmacometrics Syst Pharmacol 2022; 11:919-933. [PMID: 35570332 PMCID: PMC9286720 DOI: 10.1002/psp4.12807] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/05/2022] [Accepted: 04/13/2022] [Indexed: 11/11/2022] Open
Affiliation(s)
- Ryuta Asaumi
- Pharmacokinetic Research Laboratories Ono Pharmaceutical Co., Ltd. Ibaraki Japan
| | - Ken‐ichi Nunoya
- Pharmacokinetic Research Laboratories Ono Pharmaceutical Co., Ltd. Ibaraki Japan
| | - Yoshiyuki Yamaura
- Pharmacokinetic Research Laboratories Ono Pharmaceutical Co., Ltd. Ibaraki Japan
| | - Kunal S. Taskar
- Drug Metabolism and Pharmacokinetics In Vitro In Vivo Translation GlaxoSmithKline R&D Stevenage UK
| | - Yuichi Sugiyama
- Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, School of Pharmacy Josai International University Tokyo Japan
| |
Collapse
|
11
|
Mochizuki T, Zamek-Gliszczynski MJ, Yoshida K, Mao J, Taskar K, Hirabayashi H, Chu X, Lai Y, Takashima T, Rockich K, Yamaura Y, Fujiwara K, Mizuno T, Maeda K, Furihata K, Sugiyama Y, Kusuhara H. Effect of Cyclosporin A and Impact of Dose Staggering on OATP1B1/1B3 Endogenous Substrates and Drug Probes for Assessing Clinical Drug Interactions. Clin Pharmacol Ther 2022; 111:1315-1323. [PMID: 35292967 PMCID: PMC9325410 DOI: 10.1002/cpt.2584] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 02/28/2022] [Indexed: 12/22/2022]
Abstract
This study was designed to assess the quantitative performance of endogenous biomarkers for organic anion transporting polypeptide (OATP) 1B1/1B3‐mediated drug‐drug interactions (DDIs). Ten healthy volunteers orally received OATP1B1/1B3 probe cocktail (0.2 mg pitavastatin, 1 mg rosuvastatin, and 2 mg valsartan) and an oral dose of cyclosporin A (CysA, 20 mg and 75 mg) separated by a 1‐hour interval (20 mg (−1 hour), and 75 mg (−1 hour)). CysA 75 mg was also given with a 3‐hour interval (75 mg (−3 hours)) to examine the persistence of OATP1B1/1B3 inhibition. The area under the plasma concentration‐time curve ratios (AUCRs) were 1.63, 3.46, and 2.38 (pitavastatin), 1.39, 2.16, and 1.81 (rosuvastatin), and 1.42, 1.77, and 1.85 (valsartan), at 20 mg, 75 mg (−1 hour) and 75 mg (−3 hours) of CysA, respectively. CysA effect on OATP1B1/1B3 was unlikely to persist at the dose examined. Among 26 putative OATP1B1/1B3 biomarkers evaluated, AUCR and maximum concentration ratio (CmaxR) of CP‐I showed the highest Pearson’s correlation coefficient with CysA AUC (0.94 and 0.93, respectively). Correlation between AUCR of pitavastatin, and CmaxR or AUCR of CP‐I were consistent between this study and our previous study using rifampicin as an OATP1B1/1B3 inhibitor. Nonlinear regression analysis of AUCR−1 of pitavastatin and CP‐I against CysA Cmax yielded Ki,OATP1B1/1B3,app (109 ± 35 and 176 ± 42 nM, respectively), similar to the Ki,OATP1B1/1B3 estimated by our physiologically‐based pharmacokinetic model analysis described previously (107 nM). The endogenous OATP1B1/1B3 biomarkers, particularly CmaxR and AUCR of CP‐I, corroborates OATP1B1/1B3 inhibition and yields valuable information that improve accurate DDI predictions in drug development, and enhance our understanding of interindividual variability in the magnitude of DDIs.
Collapse
Affiliation(s)
- Tatsuki Mochizuki
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | | | - Kenta Yoshida
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Jialin Mao
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California, USA
| | - Kunal Taskar
- Drug Metabolism and Disposition, GlaxoSmithKline, Stevenage, UK
| | - Hideki Hirabayashi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | | | - Yurong Lai
- Drug Metabolism Department, Gilead Sciences Inc., Foster City, California, USA
| | - Tadayuki Takashima
- Laboratory for Safety Assessment & ADME, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Shizuoka, Japan
| | - Kevin Rockich
- Drug Metabolism, Pharmacokinetics and Clinical Pharmacology, Incyte Research Institute, Wilmington, Delaware, USA
| | - Yoshiyuki Yamaura
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd, Osaka, Japan
| | - Kaku Fujiwara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Tadahaya Mizuno
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | | | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, Technology and Innovation Hub, RIKEN, Yokohama, Kanagawa, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
12
|
Mochizuki T, Aoki Y, Yoshikado T, Yoshida K, Lai Y, Hirabayashi H, Yamaura Y, Rockich K, Taskar K, Takashima T, Chu X, Zamek-Gliszczynski MJ, Mao J, Maeda K, Furihata K, Sugiyama Y, Kusuhara H. Physiologically-based pharmacokinetic model-based translation of OATP1B-mediated drug-drug interactions from coproporphyrin I to probe drugs. Clin Transl Sci 2022; 15:1519-1531. [PMID: 35421902 DOI: 10.1111/cts.13272] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/08/2022] [Accepted: 02/13/2022] [Indexed: 11/28/2022] Open
Abstract
The accurate prediction of OATP1B-mediated drug-drug interactions (DDIs) is challenging for drug development. Here, we report physiologically-based pharmacokinetic (PBPK) model analysis for clinical DDI data generated in heathy subjects who received oral doses of cyclosporin A (CysA; 20 and 75 mg) as an OATP1B inhibitor, and the probe drugs (pitavastatin, rosuvastatin and valsartan). PBPK models of CysA and probe compounds were combined assuming inhibition of hepatic uptake of endogenous coproporphyrin I (CP-I) by CysA. In vivo Ki of unbound CysA for OATP1B (Ki,OATP1B ), and the overall intrinsic hepatic clearance per body weight of CP-I (CLint,all,unit ) were optimized to account for the CP-I data (Ki,OATP1B , 0.657 ± 0.048 nM; CLint,all,unit , 57.0 ± 6.3 L/h/kg). DDI simulation using Ki,OATP1B reproduced the dose-dependent effect of CysA (20 and 75 mg) and the dosing interval (1 h and 3 h) on the time profiles of blood concentrations of pitavastatin and rosuvastatin, but DDI simulation using in vitro Ki,OATP1B failed. The Cluster Gauss-Newton method was used to conduct parameter optimization using 1,000 initial parameter sets for the seven pharmacokinetic parameters of CP-I (β, CLint,all , Fa Fg , Rdif , fbile , fsyn , and vsyn ), and Ki,OATP1B , and Ki,MRP2 of CysA. Based on the accepted 498 parameter sets, the range of CLint,all and Ki,OATP1B was narrowed, with coefficients of variation (CVs) of 9.3% and 11.1%, respectively, indicating that these parameters were practically identifiable. These results suggest that PBPK model analysis of CP-I is a promising translational approach to predict OATP1B-mediated DDIs in drug development.
Collapse
Affiliation(s)
- Tatsuki Mochizuki
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo
| | - Yasunori Aoki
- Laboratory of quantitative system pharmacokinetics / pharmacodynamics, Josai International University, School of Pharmacy, Tokyo, Japan
| | - Takashi Yoshikado
- Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Yokohama, Kanagawa, Japan
| | - Kenta Yoshida
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, California, USA
| | - Hideki Hirabayashi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | - Yoshiyuki Yamaura
- Pharmacokinetic Research Laboratories , Ono Pharmaceutical Co., Ltd., Osaka, Japan
| | - Kevin Rockich
- Drug Metabolism, Pharmacokinetics and Clinical Pharmacology, Incyte Research Institute, Wilmington, Delaware, USA
| | - Kunal Taskar
- Drug Metabolism and Pharmacokinetics, IVIVT, GlaxoSmithKline, Stevenage, UK
| | - Tadayuki Takashima
- Laboratory for Safety Assessment & ADME, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Shizuoka, Japan
| | - Xiaoyan Chu
- Department of Pharmacokinetics, Pharmacodynamics & Drug Metabolism, Merck & Co., Inc., Kenilworth, NJ, USA
| | | | - Jialin Mao
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California, USA
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo
| | | | - Yuichi Sugiyama
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo.,Laboratory of quantitative system pharmacokinetics / pharmacodynamics, Josai International University, School of Pharmacy, Tokyo, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, the University of Tokyo
| |
Collapse
|
13
|
Izumi S, Nozaki Y, Lee W, Sugiyama Y. Experimental and modeling evidence supporting the trans-inhibition mechanism for preincubation time-dependent, long-lasting inhibition of organic anion transporting polypeptide (OATP) 1B1 by cyclosporine A. Drug Metab Dispos 2022; 50:541-551. [PMID: 35241487 DOI: 10.1124/dmd.121.000783] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/14/2022] [Indexed: 11/22/2022] Open
Abstract
Cyclosporine A (CsA) and rifampin are potent inhibitors of organic anion transporting polypeptide (OATP) 1B1 and are widely used to assess the risk for drug-drug interactions. CsA displays preincubation time-dependent, long-lasting inhibition of OATP1B1 in vitro and in rats in vivo, and a proposed mechanism is the trans-inhibition by which CsA inhibits OATP1B1 from the inside of cells. The current study aimed to experimentally validate the proposed mechanism using HEK293 cells stably expressing OATP1B1. The uptake of CsA reached a plateau following around 60-min incubation, with the cell-to-buffer concentration ratio of 3930, reflective of the high-affinity, high-capacity intracellular binding of CsA. The time course of CsA uptake was analyzed to estimate the kinetic parameters for permeability clearance and intracellular binding. When the OATP1B1-mediated uptake of [3H]estradiol-17β-glucuronide was measured following preincubation with CsA for 5 to 120 min, apparent Ki values became lower with longer preincubation. Our kinetic modeling incorporated the two reversible inhibition constants [Ki,trans and Ki,cis for the inhibition from inside (trans-inhibition) and outside (cis-inhibition) of cells, respectively] and estimated Ki,trans value of CsA was smaller by 48-fold than the estimated Ki,cis value. Rifampin also displayed preincubation time-dependent inhibition of OATP1B1, albeit the extent of enhancement was only 2-fold. The current study provides experimental evidence for the preincubation time-dependent shift of apparent Ki values and a mechanistic basis for physiologically based pharmacokinetic modeling that incorporates permeability clearance, extensive intracellular binding, and asymmetry of Ki values between the inside and outside of cells. Significance Statement In vitro data and kinetic modeling support that preincubation time-dependent, long-lasting inhibition of OATP1B1 by CsA can be explained by the extensive intracellular binding and reversible OATP1B1 inhibition intracellularly (trans-inhibition) as well as extracellularly (cis-inhibition). For inhibitors to display time-dependency, the following factors were found important: time to reach a steady-state cellular concentration, trans-inhibition potency relative to cis-inhibition, and the degree of cellular inhibitor accumulation. This study would aid in the accurate prediction of drug-drug interactions mediated by OATP1B1 inhibition.
Collapse
Affiliation(s)
| | | | - Wooin Lee
- Seoul National University, Korea, Republic of
| | - Yuichi Sugiyama
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| |
Collapse
|
14
|
Konno S, Kobayashi K, Senda M, Funai Y, Seki Y, Tamai I, Schäkel L, Sakata K, Pillaiyar T, Taguchi A, Taniguchi A, Gütschow M, Müller CE, Takeuchi K, Hirohama M, Kawaguchi A, Kojima M, Senda T, Shirasaka Y, Kamitani W, Hayashi Y. 3CL Protease Inhibitors with an Electrophilic Arylketone Moiety as Anti-SARS-CoV-2 Agents. J Med Chem 2022; 65:2926-2939. [PMID: 34313428 PMCID: PMC8340582 DOI: 10.1021/acs.jmedchem.1c00665] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Indexed: 02/08/2023]
Abstract
The novel coronavirus, SARS-CoV-2, has been identified as the causative agent for the current coronavirus disease (COVID-19) pandemic. 3CL protease (3CLpro) plays a pivotal role in the processing of viral polyproteins. We report peptidomimetic compounds with a unique benzothiazolyl ketone as a warhead group, which display potent activity against SARS-CoV-2 3CLpro. The most potent inhibitor YH-53 can strongly block the SARS-CoV-2 replication. X-ray structural analysis revealed that YH-53 establishes multiple hydrogen bond interactions with backbone amino acids and a covalent bond with the active site of 3CLpro. Further results from computational and experimental studies, including an in vitro absorption, distribution, metabolism, and excretion profile, in vivo pharmacokinetics, and metabolic analysis of YH-53 suggest that it has a high potential as a lead candidate to compete with COVID-19.
Collapse
Affiliation(s)
- Sho Konno
- School of Pharmacy, Department of Medicinal Chemistry,
Tokyo University of Pharmacy and Life Sciences, Hachioji,
Tokyo, 192-0392, Japan
| | - Kiyotaka Kobayashi
- School of Pharmacy, Department of Medicinal Chemistry,
Tokyo University of Pharmacy and Life Sciences, Hachioji,
Tokyo, 192-0392, Japan
| | - Miki Senda
- Structural Biology Research Center, Institute of
Materials Structure Science, High Energy Accelerator Research Organization
(KEK), Tsukuba 305-0801, Japan
| | - Yuta Funai
- Faculty of Pharmacy, Institute of Medical,
Pharmaceutical and Health Sciences, Kanazawa University,
Kanazawa 920-1192, Japan
| | - Yuta Seki
- Faculty of Pharmacy, Institute of Medical,
Pharmaceutical and Health Sciences, Kanazawa University,
Kanazawa 920-1192, Japan
| | - Ikumi Tamai
- Faculty of Pharmacy, Institute of Medical,
Pharmaceutical and Health Sciences, Kanazawa University,
Kanazawa 920-1192, Japan
| | - Laura Schäkel
- Pharmaceutical Institute, Pharmaceutical &
Medicinal Chemistry, University of Bonn, Bonn 53121,
Germany
| | - Kyousuke Sakata
- School of Life Sciences, Tokyo University
of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392,
Japan
| | - Thanigaimalai Pillaiyar
- Pharmaceutical Institute, Pharmaceutical/Medicinal Chemistry,
University of Tübingen, Tübingen 72076,
Germany
| | - Akihiro Taguchi
- School of Pharmacy, Department of Medicinal Chemistry,
Tokyo University of Pharmacy and Life Sciences, Hachioji,
Tokyo, 192-0392, Japan
| | - Atsuhiko Taniguchi
- School of Pharmacy, Department of Medicinal Chemistry,
Tokyo University of Pharmacy and Life Sciences, Hachioji,
Tokyo, 192-0392, Japan
| | - Michael Gütschow
- Pharmaceutical Institute, Pharmaceutical &
Medicinal Chemistry, University of Bonn, Bonn 53121,
Germany
| | - Christa E. Müller
- Pharmaceutical Institute, Pharmaceutical &
Medicinal Chemistry, University of Bonn, Bonn 53121,
Germany
| | - Koh Takeuchi
- Cellular and Molecular Biotechnology Research
Institute, National Institute of Advanced Industrial Science and
Technology, Koto, Tokyo 135-0064, Japan
| | - Mikako Hirohama
- Faculty of Medicine, Transborder Medical Research
Center, University of Tsukuba, Tsukuba 305-8575,
Japan
| | - Atsushi Kawaguchi
- Faculty of Medicine, Transborder Medical Research
Center, University of Tsukuba, Tsukuba 305-8575,
Japan
| | - Masaki Kojima
- School of Life Sciences, Tokyo University
of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392,
Japan
| | - Toshiya Senda
- Structural Biology Research Center, Institute of
Materials Structure Science, High Energy Accelerator Research Organization
(KEK), Tsukuba 305-0801, Japan
| | - Yoshiyuki Shirasaka
- Faculty of Pharmacy, Institute of Medical,
Pharmaceutical and Health Sciences, Kanazawa University,
Kanazawa 920-1192, Japan
| | - Wataru Kamitani
- Department of Infectious Diseases and Host Defense,
Gunma University Graduate School of Medicine, Maebashi
371-8511, Japan
| | - Yoshio Hayashi
- School of Pharmacy, Department of Medicinal Chemistry,
Tokyo University of Pharmacy and Life Sciences, Hachioji,
Tokyo, 192-0392, Japan
| |
Collapse
|
15
|
Suzuki S, Inoue K, Tamai I, Shirasaka Y. Model Analysis of the Apparent Saturation Kinetics of Purine Nucleobase Uptake in Cells co-Expressing Transporter and Metabolic Enzyme. Pharm Res 2021; 38:1585-1592. [PMID: 34435306 DOI: 10.1007/s11095-021-03086-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/18/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE This study aims to understand the effect of salvage enzyme activity on the saturable kinetics of facilitated cellular uptake of purine nucleobase by developing a cellular kinetic model incorporating equilibrative nucleobase transporter 1 (ENBT1) and adenine phosphoribosyltransferase (APRT), with adenine as a model nucleobase. METHODS A cellular kinetic model incorporating the functions of ENBT1 and APRT was developed using Napp software and employed for model-based analysis of the cellular disposition of adenine. RESULTS Simulation analysis using the developed cellular kinetic model could account for the experimentally observed time-dependent changes in the Km(app) value of adenine for ENBT1-mediated uptake. At a long experimental time, the model shows that uptake of adenine is rate-limited by APRT, enabling determination of the Km value for APRT. At early time, the rate-limiting step for adenine uptake is ENBT1-mediated transport, enabling determination of the Km value for ENBT1. Further simulations showed that the effect of experimental time on the Km(app) value for ENBT1-mediated uptake is dependent on the APRT expression level. CONCLUSION Our findings indicate that both enzyme expression levels and experimental time should be considered when using cellular uptake studies to determine the Km values of purine nucleobases for facilitated transporters.
Collapse
Affiliation(s)
- Satoru Suzuki
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan.,School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Tokyo, 192-0392, Japan
| | - Katsuhisa Inoue
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Tokyo, 192-0392, Japan
| | - Ikumi Tamai
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Yoshiyuki Shirasaka
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan. .,School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Tokyo, 192-0392, Japan.
| |
Collapse
|
16
|
Asano S, Yoshitomo A, Hozuki S, Sato H, Kazuki Y, Hisaka A. A New Intestinal Model for Analysis of Drug Absorption and Interactions Considering Physiological Translocation of Contents. Drug Metab Dispos 2021; 49:581-591. [PMID: 33962977 DOI: 10.1124/dmd.121.000361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/09/2021] [Indexed: 11/22/2022] Open
Abstract
Precise prediction of drug absorption is key to the success of new drug development and efficacious pharmacotherapy. In this study, we developed a new absorption model, the advanced translocation model (ATOM), by extending our previous model, the translocation model. ATOM reproduces the translocation of a substance in the intestinal lumen using a partial differential equation with variable dispersion and convection terms to describe natural flow and micromixing within the intestine under not only fasted but also fed conditions. In comparison with ATOM, it was suggested that a conventional absorption model, advanced compartmental absorption and transit model, tends to underestimate micromixing in the upper intestine, and it is difficult to adequately describe movements under the fasted and fed conditions. ATOM explains the observed nonlinear absorption of midazolam successfully, with a minimal number of scaling factors. Furthermore, ATOM considers the apical and basolateral membrane permeabilities of enterocytes separately and assumes compartmentation of the lamina propria, including blood vessels, to consider intestinal blood flow appropriately. ATOM estimates changes in the intestinal availability caused by drug interaction associated with inhibition of CYP3A and P-glycoprotein in the intestine. Additionally, ATOM can estimate the drug absorption in the fed state considering delayed intestinal drug flow. Therefore, ATOM is a useful tool for the analysis of local pharmacokinetics in the gastrointestinal tract, especially for the estimation of nonlinear drug absorption, which may involve various interactions with intestinal contents or other drugs. SIGNIFICANCE STATEMENT: The newly developed advanced translocation model precisely explains various movements of intestinal contents under fasted and fed conditions, which cannot be adequately described by the current physiological pharmacokinetic models.
Collapse
Affiliation(s)
- Satoshi Asano
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (S.A., A.Y., S.H., H.S., A.H.); DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A.); Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Aoi Yoshitomo
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (S.A., A.Y., S.H., H.S., A.H.); DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A.); Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Shizuka Hozuki
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (S.A., A.Y., S.H., H.S., A.H.); DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A.); Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Hiromi Sato
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (S.A., A.Y., S.H., H.S., A.H.); DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A.); Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Yasuhiro Kazuki
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (S.A., A.Y., S.H., H.S., A.H.); DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A.); Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| | - Akihiro Hisaka
- Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan (S.A., A.Y., S.H., H.S., A.H.); DMPK Research Department, Teijin Pharma Limited, Tokyo, Japan (S.A.); Chromosome Engineering Research Center (Y.K.) and Division of Genome and Cellular Functions, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine (Y.K.), Tottori University, Tottori, Japan
| |
Collapse
|
17
|
Ochiai W, Kitaoka S, Kawamura T, Hatogai J, Harada S, Iizuka M, Ariumi M, Takano S, Nagai T, Sasatsu M, Sugiyama K. Maternal and Fetal Pharmacokinetic Analysis of Cannabidiol during Pregnancy in Mice. Drug Metab Dispos 2021; 49:337-343. [PMID: 33531413 DOI: 10.1124/dmd.120.000270] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/25/2021] [Indexed: 11/22/2022] Open
Abstract
Cannabidiol (CBD), a major component of cannabis, has various effects, such as antiemetic and anxiolytic activities, and has recently been marketed as a supplement. The number of people using CBD during pregnancy is increasing, and there are concerns about its effects on the fetus. In addition, the scientific evidence supporting the fetal safety of CBD use during pregnancy is insufficient. To investigate CBD transfer from the mother to the fetus, a single intravenous dose of CBD was administered to pregnant mice in this study, and fetal pharmacokinetics (distribution and elimination) was analyzed. The transfer of CBD from the maternal blood to the fetus was rapid, and the compound accumulated in the fetal brain, liver, and gastrointestinal tract. Conversely, little CBD was transferred from the mother to the amniotic fluid. We analyzed the pharmacokinetics of CBD using a two-compartment model and found that the maternal and fetal half-lives of CBD were approximately 5 and 2 hours, respectively. Furthermore, we performed a moment analysis of the pharmacokinetics of CBD, observing a mean residence time of less than 2 hours in both the mother and fetus. These results suggest that once-daily CBD intake during pregnancy is unlikely to result in CBD accumulation in the mother or fetus. SIGNIFICANCE STATEMENT: CBD is currently marketed as a supplement, and despite its increasing use during pregnancy, little information concerning its fetal effects has been reported. In the present study, CBD was administered to pregnant mice, and the pharmacokinetics in the fetus was investigated using a two-compartment model and moment analysis. The results of these analyses provide important information for estimating the risk to the fetus if CBD is mistakenly consumed during pregnancy.
Collapse
Affiliation(s)
- Wataru Ochiai
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Satoshi Kitaoka
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Taisuke Kawamura
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Jo Hatogai
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Shohei Harada
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Misa Iizuka
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Mashu Ariumi
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Seiya Takano
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Tomomi Nagai
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Masanaho Sasatsu
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Kiyoshi Sugiyama
- Department of Pharmacokinetics (W.O., S.K., T.K., J.H., S.H., M.I., M.A., S.T., T.N.), Laboratory of Tissue Regeneration (M.S.), and Department of Functional Molecule Kinetics (K.S.), School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan
| |
Collapse
|
18
|
Hotta T, Tsubata Y, Hamai K, Tanino A, Kobayashi M, Nakamura A, Sugisaka J, Hongoh M, Ishihara N, Ishikawa N, Yamasaki M, Fujitaka K, Kubota T, Nishimura N, Isobe T. Pharmacokinetics of edoxaban in EGFR-mutated non-small cell lung cancer patients with venous thromboembolism. Respir Investig 2021; 59:327-334. [PMID: 33386292 DOI: 10.1016/j.resinv.2020.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/14/2020] [Accepted: 11/30/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND The risk of venous thromboembolism (VTE) is increased 7-fold in patients with cancer than in those without. Low-molecular-weight heparin is the standard treatment for cancer-associated VTE. Direct oral anticoagulants (DOACs) are not inferior to low-molecular-weight heparin with respect to the general outcome of recurrent VTE. Warfarin is associated with a risk of bleeding when used in combination with gefitinib or erlotinib which are epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs). It is unclear, however, whether combination treatments with EGFR-TKIs and DOACs pose the same risk. We aimed to identify anticancer drugs and anticoagulants that can be used safely in combination, as accompanying research to an observational study on VTE incidence rates in lung cancer patients (Rising-VTE/NEJ037 study). METHODS Twelve patients receiving EFGR-TKI monotherapy and VTE treatment were enrolled. Blood samples were collected in time series after the first dose of edoxaban, and further samples were collected within 8-15 days after administering EGFR-TKIs. The pharmacokinetics (PK) of edoxaban were analyzed using a non-compartmental model. RESULTS Edoxaban concentrations (30 mg once daily) were measured in eight patients. PK analyses showed no significant differences before and after co-administration of EGFR-TKIs (gefitinib, erlotinib, and afatinib). CONCLUSIONS Our findings indicate that the PK of edoxaban was not considerably affected by co-administration of EGFR-TKIs (gefitinib, erlotinib, and afatinib).
Collapse
Affiliation(s)
- Takamasa Hotta
- Department of Internal Medicine, Division of Medical Oncology & Respiratory Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Yukari Tsubata
- Department of Internal Medicine, Division of Medical Oncology & Respiratory Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan.
| | - Kosuke Hamai
- Department of Respiratory Medicine, Hiroshima Prefectural Hospital, 1-5-54 Ujina-Kanda, Minami-ku, Hiroshima, 734-8530, Japan
| | - Akari Tanino
- Department of Internal Medicine, Division of Medical Oncology & Respiratory Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Misato Kobayashi
- Department of Internal Medicine, Division of Medical Oncology & Respiratory Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Atsushi Nakamura
- Department of Pulmonary Medicine, Sendai Kousei Hospital, 4-15 Aoba-ku, Sendai, Miyagi, 980-0873, Japan
| | - Jun Sugisaka
- Department of Pulmonary Medicine, Sendai Kousei Hospital, 4-15 Aoba-ku, Sendai, Miyagi, 980-0873, Japan
| | - Masafumi Hongoh
- Department of Pharmacy, Shimane University Hospital, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Noriyuki Ishihara
- Department of Pharmacy, Shimane University Hospital, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Nobuhisa Ishikawa
- Department of Respiratory Medicine, Hiroshima Prefectural Hospital, 1-5-54 Ujina-Kanda, Minami-ku, Hiroshima, 734-8530, Japan
| | - Masahiro Yamasaki
- Department of Respiratory Disease, Hiroshima Red Cross Hospital & Atomic-Bomb Survivors Hospital, 1-9-6 Naka-ku, Hiroshima, 730-8619, Japan
| | - Kazunori Fujitaka
- Department of Respiratory Internal Medicine, Hiroshima University Hospital, Kasumi 1-2-3, Minami-ku, Hiroshima, 734-8551, Japan
| | - Tetsuya Kubota
- Department of Respiratory Medicine and Allergology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku-shi, Kochi, 783-8505, Japan
| | - Nobuhiro Nishimura
- Department of Pharmacology, School of Pharmaceutical Sciences at Fukuoka International University of Health and Welfare, 137-1 Enokizu, Okawa, Fukuoka, 831-8501, Japan
| | - Takeshi Isobe
- Department of Internal Medicine, Division of Medical Oncology & Respiratory Medicine, Shimane University, 89-1 Enya-cho, Izumo, Shimane, 693-8501, Japan
| |
Collapse
|
19
|
Yoshikado T, Lee W, Toshimoto K, Morita K, Kiriake A, Chu X, Lee N, Kimoto E, Varma MVS, Kikuchi R, Scialis RJ, Shen H, Ishiguro N, Lotz R, Li AP, Maeda K, Kusuhara H, Sugiyama Y. Evaluation of Hepatic Uptake of OATP1B Substrates by Short Term-Cultured Plated Human Hepatocytes: Comparison With Isolated Suspended Hepatocytes. J Pharm Sci 2020; 110:376-387. [PMID: 33122051 DOI: 10.1016/j.xphs.2020.10.041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/13/2022]
Abstract
Hepatic uptake clearance has been measured in suspended human hepatocytes (SHH). Plated human hepatocytes (PHH) after short-term culturing are increasingly employed to study hepatic transport driven mainly by its higher throughput. To know pros/cons of both systems, the hepatic uptake clearances of several organic anion transporting polypeptide 1B substrates were compared between PHH and SHH by determining the initial uptake velocities or through dynamic model-based analyses. For cerivastatin, pitavastatin and rosuvastatin, initial uptake clearances (PSinf) obtained using PHH were comparable to those using SHH, while cell-to-medium concentration (C/M) ratios were 2.7- to 5.4-fold higher. For pravastatin and dehydropravastatin, hydrophilic compounds with low uptake/cellular binding, their PSinf and C/M ratio in PHH were 1.8- to 3.2-fold lower than those in SHH. These hydrophilic substrates are more prone to wash-off during the uptake study using PHH, which may explain the apparently lower uptake than SHH. The C/M ratios obtained using PHH were stable over an extended time, making PHH suitable to estimate the C/M ratios and hepatocyte-to-medium unbound concentration ratios (Kp,uu). In conclusion, PHH is useful in evaluating hepatic uptake/efflux clearances and Kp,uu of OATP1B substrates in a high-throughput manner, however, a caution is warranted for hydrophilic drugs with low uptake/cellular binding.
Collapse
Affiliation(s)
- Takashi Yoshikado
- Sugiyama Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, Technology and Innovation Hub, RIKEN, Yokohama, Kanagawa, Japan; Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Yokohama, Kanagawa, Japan
| | - Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, Korea
| | - Kota Toshimoto
- Sugiyama Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, Technology and Innovation Hub, RIKEN, Yokohama, Kanagawa, Japan
| | - Kiyoe Morita
- Sugiyama Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, Technology and Innovation Hub, RIKEN, Yokohama, Kanagawa, Japan
| | - Aya Kiriake
- Sugiyama Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, Technology and Innovation Hub, RIKEN, Yokohama, Kanagawa, Japan
| | | | - Nora Lee
- Daewoong Pharmaceutical Co., Ltd, Seoul, Korea
| | - Emi Kimoto
- ADME Sciences, Medicine Design, Worldwide Research and Development, Pfizer Inc, Groton, CT, USA
| | - Manthena V S Varma
- ADME Sciences, Medicine Design, Worldwide Research and Development, Pfizer Inc, Groton, CT, USA
| | | | | | - Hong Shen
- Bristol Myers Squibb, Princeton, NJ, USA
| | - Naoki Ishiguro
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd, Kobe, Hyogo, Japan
| | - Ralf Lotz
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co., KG, Biberach an der Riss, Germany
| | - Albert P Li
- In Vitro ADMET Laboratories Inc, Columbia, MA, USA
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, Technology and Innovation Hub, RIKEN, Yokohama, Kanagawa, Japan.
| |
Collapse
|
20
|
Lee W, Koyama S, Morita K, Kiriake A, Kikuchi R, Chu X, Lee N, Scialis RJ, Shen H, Kimoto E, Tremaine L, Ishiguro N, Lotz R, Maeda K, Kusuhara H, Sugiyama Y. Cell-to-Medium Concentration Ratio Overshoot in the Uptake of Statins by Human Hepatocytes in Suspension, but Not in Monolayer: Kinetic Analysis Suggesting a Partial Loss of Functional OATP1Bs. AAPS JOURNAL 2020; 22:133. [PMID: 33063163 PMCID: PMC7561564 DOI: 10.1208/s12248-020-00512-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/15/2020] [Indexed: 01/16/2023]
Abstract
Suspended human hepatocytes (SHH) have long been used in assessing hepatic drug uptake, while plated human hepatocytes in short-term monolayer culture (PHH) have gained use in recent years. This study aimed to cross-evaluate SHH and PHH in measuring the hepatic uptake mediated by organic anion transporting polypeptide 1Bs (OATP1Bs). We compared the time courses of cell-to-medium (C/M) concentration ratios and initial uptake clearance values of the OATP1B substrates (pitavastatin, rosuvastatin, cerivastatin, pravastatin, dehydropravastatin, and SC-62807) between SHH and PHH. For all compounds except cerivastatin, the C/M ratios in SHH displayed an apparent overshoot (an initial increase followed by a decrease) during the 180-min uptake experiment, but not in PHH. Based on the literature evidence suggesting the possible internalization of OATP1Bs in primary hepatocytes, separate experiments measured the drug uptake after varying lengths of pre-incubation in the drug-free medium. The initial uptake clearances of pitavastatin and rosuvastatin declined in SHH beyond an apparent threshold time of 20-min drug-free pre-incubation, but not in PHH. Kinetic modeling quantitatively captured the decline in the active uptake clearance in SHH, and more than half of the active uptake clearances of pitavastatin and rosuvastatin were prone to loss during the 180-min uptake experiment. These results suggested a partial, time-delayed loss of the functional OATP1Bs in SHH upon prolonged incubation. Our results indicate that PHH is more appropriate for experiments where a prolonged incubation is required, such as estimation of unbound hepatocyte-to-medium concentration ratio (Kp,uu) at the steady-state.
Collapse
Affiliation(s)
- Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, South Korea
| | - Satoshi Koyama
- Sugiyama Laboratory, RIKEN Cluster for Science, Technology and Innovation Hub, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, Japan
| | - Kiyoe Morita
- Sugiyama Laboratory, RIKEN Cluster for Science, Technology and Innovation Hub, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, Japan
| | - Aya Kiriake
- Sugiyama Laboratory, RIKEN Cluster for Science, Technology and Innovation Hub, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, Japan
| | | | - Xiaoyan Chu
- Merck & Co., Inc, North Wales, Pennsylvania, USA
| | - Nora Lee
- Daewoong Pharmaceutical Co., Ltd, Seoul, South Korea
| | | | - Hong Shen
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Emi Kimoto
- ADME Sciences, Medicine Design, Worldwide Research and Development, Pfizer Inc, Groton, Connecticut, USA
| | - Larry Tremaine
- Tremaine DMPK Consulting LLC, Merritt Island, Florida, USA
| | - Naoki Ishiguro
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd, Kobe, Hyogo, Japan
| | - Ralf Lotz
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Cluster for Science, Technology and Innovation Hub, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, Japan.
| |
Collapse
|
21
|
Jin R, Hisaka A. Implementing PRED Subroutine of NONMEM for Versatile Pharmacokinetic Analysis Using Fast Inversion of Laplace Transform (FILT). Chem Pharm Bull (Tokyo) 2020; 68:891-894. [PMID: 32611991 DOI: 10.1248/cpb.c20-00236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In pharmacokinetic (PK) analysis, conventional models are described by ordinary differential equations (ODE) that are generally solved in their Laplace transformed forms. The solution in the Laplace transformed forms is inverse Laplace transformed to derive an analytical solution. However, inverse Laplace transform is often mathematically difficult. Consequently, numerical inverse Laplace transform methods have been developed. In this study, we focus on extending the modeling functions of Nonlinear Mixed Effect Model (NONMEM), a standard software for PK and population pharmacokinetic (PPK) analyses, by adding the Fast Inversion of Laplace Transform (FILT) method, one of the representative numerical inverse Laplace transform methods. We implemented PREDFILT, a specialized PRED subroutine, which functions as an internal model unit in NONMEM to enable versatile FILT analysis with second-order precision. The calculation results of the compartment models and a dispersion model are in good agreement with the ordinary analytical solutions and theoretical values. Therefore, PREDFILT ensures enhanced flexibility in PK or PPK analyses under NONMEM environments.
Collapse
Affiliation(s)
- Ryota Jin
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University
| | - Akihiro Hisaka
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University
| |
Collapse
|
22
|
Lack of Drug-Drug Interaction Between Cimetidine, a Renal Transporter Inhibitor, and Imeglimin, a Novel Oral Antidiabetic Drug, in Healthy Volunteers. Eur J Drug Metab Pharmacokinet 2020; 45:725-733. [PMID: 32860624 DOI: 10.1007/s13318-020-00642-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND AND OBJECTIVE: Imeglimin is a novel oral antidiabetic drug to treat type 2 diabetes, targeting the mitochondrial bioenergetics. In vitro, imeglimin was shown to be a substrate of human multidrug and toxic extrusion transporters MATE1 and MATE2-K and organic cation transporters OCT1 and OCT2. The objective of the study was to assess the potential drug-drug interaction between imeglimin and cimetidine, a reference inhibitor of these transporters. METHODS A phase 1 study was carried out in 16 subjects who received a single dose of 1500 mg imeglimin alone on day 1 followed by a 6-day treatment (day 5 to day 10) with cimetidine 400 mg twice daily. On day 8, a single dose of imeglimin was co-administered with cimetidine. Blood and urine samples were collected up to 72 h after each imeglimin administration. Pharmacokinetic parameters were determined using non-compartmental methods. RESULTS Imeglimin maximum plasma concentration (Cmax) and area under the plasma concentration-time curve (AUC) were 1.3-fold [90% CI (1.12-1.62) and (1.10-1.46) for Cmax and AUC0-last, respectively] higher when imeglimin was co-administered with cimetidine but this increase was not considered clinically relevant. This increase could be mainly explained by a reduction in renal elimination, mediated through the cimetidine inhibition of renal MATE1 transporter. Imeglimin taken alone or with cimetidine was safe and well tolerated in all subjects. CONCLUSIONS No clinically significant drug-drug interaction exists between imeglimin and cimetidine, a reference inhibitor of MATE1, MATE2-K, OCT1 and OCT2 transporters. CLINICAL TRIAL REGISTRATION EudraCT 2018-001103-36.
Collapse
|
23
|
Wada S, Matsunaga N, Tamai I. Mathematical modeling analysis of hepatic uric acid disposition using human sandwich-cultured hepatocytes. Drug Metab Pharmacokinet 2020; 35:432-440. [PMID: 32807664 DOI: 10.1016/j.dmpk.2020.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 05/24/2020] [Accepted: 06/15/2020] [Indexed: 12/14/2022]
Abstract
Uric acid is biosynthesized from purine by xanthine oxidase (XO) mainly in the liver and is excreted into urine and feces. Although several transporters responsible for renal and intestinal handling of uric acid have been reported, information on hepatic transporters is limited. In the present study, we studied quantitative contribution of transporters for hepatic handling of uric acid by mathematical modeling analysis in human sandwich-cultured hepatocytes (hSCH). Stable isotope-labeled hypoxanthine, hypoxanthine-13C2,15N (HX), was incubated with hSCH and formed 13C2,15N-labeled xanthine (XA) and uric acid (UA) were measured by LC-MS/MS time dependently. Rate constants for metabolism and efflux and uptake transport across sinusoidal and bile canalicular membranes of HX, XA and UA were estimated in the presence of inhibitors of XO and uric acid transporters. An XO inhibitor allopurinol significantly decreased metabolisms of HX and XA. Efflux into bile canalicular lumen was negligible and sinusoidal efflux was considered main efflux pathway of formed UA. Transporter inhibition study highlighted that GLUT9 strongly and MRP4 intermediately contribute to the sinusoidal efflux of UA with minor contribution of NPT1/4. Modeling analysis developed in the present study should be useful for quantitative prediction of uric acid disposition in liver.
Collapse
Affiliation(s)
- Sho Wada
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Japan
| | - Norikazu Matsunaga
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Japan
| | - Ikumi Tamai
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Japan.
| |
Collapse
|
24
|
Quantitative analysis of the effect of controlled-release formulation on nonlinear gastrointestinal absorption of P-glycoprotein substrate talinolol using physiologically based pharmacokinetic absorption model. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2019.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
25
|
Lee N, Maeda K, Fukizawa S, Ieiri I, Tomaru A, Akao H, Takeda K, Iwadare M, Niwa O, Masauji T, Yamane N, Kajinami K, Kusuhara H, Sugiyama Y. Microdosing clinical study to clarify pharmacokinetic and pharmacogenetic characteristics of atorvastatin in Japanese hypercholesterolemic patients. Drug Metab Pharmacokinet 2019; 34:387-395. [DOI: 10.1016/j.dmpk.2019.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/28/2019] [Accepted: 08/13/2019] [Indexed: 10/26/2022]
|
26
|
Asaumi R, Menzel K, Lee W, Nunoya KI, Imawaka H, Kusuhara H, Sugiyama Y. Expanded Physiologically-Based Pharmacokinetic Model of Rifampicin for Predicting Interactions With Drugs and an Endogenous Biomarker via Complex Mechanisms Including Organic Anion Transporting Polypeptide 1B Induction. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2019; 8:845-857. [PMID: 31420941 PMCID: PMC6875706 DOI: 10.1002/psp4.12457] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/08/2019] [Indexed: 02/01/2023]
Abstract
As rifampicin can cause the induction and inhibition of multiple metabolizing enzymes and transporters, it has been challenging to accurately predict the complex drug–drug interactions (DDIs). We previously constructed a physiologically‐based pharmacokinetic (PBPK) model of rifampicin accounting for the components for the induction of cytochrome P450 (CYP) 3A/CYP2C9 and the inhibition of organic anion transporting polypeptide 1B (OATP1B). This study aimed to expand and verify the PBPK model for rifampicin by incorporating additional components for the induction of OATP1B and CYP2C8 and the inhibition of multidrug resistance protein 2. The established PBPK model was capable of accurately predicting complex rifampicin‐induced alterations in the profiles of glibenclamide, repaglinide, and coproporphyrin I (an endogenous biomarker of OATP1B activities) with various dosing regimens. Our comprehensive rifampicin PBPK model may enable quantitative prediction of DDIs across diverse potential victim drugs and endogenous biomarkers handled by multiple metabolizing enzymes and transporters.
Collapse
Affiliation(s)
- Ryuta Asaumi
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Tsukuba, Japan
| | | | - Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Ken-Ichi Nunoya
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Tsukuba, Japan
| | - Haruo Imawaka
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Tsukuba, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, Technology and Innovation Hub, RIKEN, Yokohama, Japan
| |
Collapse
|
27
|
Nishiyama K, Toshimoto K, Lee W, Ishiguro N, Bister B, Sugiyama Y. Physiologically-Based Pharmacokinetic Modeling Analysis for Quantitative Prediction of Renal Transporter-Mediated Interactions Between Metformin and Cimetidine. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2019; 8:396-406. [PMID: 30821133 PMCID: PMC6617824 DOI: 10.1002/psp4.12398] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/06/2019] [Indexed: 12/24/2022]
Abstract
Metformin is an important antidiabetic drug and often used as a probe for drug–drug interactions (DDIs) mediated by renal transporters. Despite evidence supporting the inhibition of multidrug and toxin extrusion proteins as the likely DDI mechanism, the previously reported physiologically‐based pharmacokinetic (PBPK) model required the substantial lowering of the inhibition constant values of cimetidine for multidrug and toxin extrusion proteins from those obtained in vitro to capture the clinical DDI data between metformin and cimetidine.1 We constructed new PBPK models in which the transporter‐mediated uptake of metformin is driven by a constant membrane potential. Our models successfully captured the clinical DDI data using in vitro inhibition constant values and supported the inhibition of multidrug and toxin extrusion proteins by cimetidine as the DDI mechanism upon sensitivity analysis and data fitting. Our refined PBPK models may facilitate prediction approaches for DDI involving metformin using in vitro inhibition constant values.
Collapse
Affiliation(s)
- Kotaro Nishiyama
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Hyogo, Japan
| | - Kota Toshimoto
- Sugiyama Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, Technology and Innovation Hub, Yokohama, Kanagawa, Japan
| | - Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Naoki Ishiguro
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Hyogo, Japan
| | - Bojan Bister
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Hyogo, Japan
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, Technology and Innovation Hub, Yokohama, Kanagawa, Japan
| |
Collapse
|
28
|
Kim SJ, Lee KR, Miyauchi S, Sugiyama Y. Extrapolation of In Vivo Hepatic Clearance from In Vitro Uptake Clearance by Suspended Human Hepatocytes for Anionic Drugs with High Binding to Human Albumin: Improvement of In Vitro-to-In Vivo Extrapolation by Considering the “Albumin-Mediated” Hepatic Uptake Mechanism on the Basis of the “Facilitated-Dissociation Model”. Drug Metab Dispos 2018; 47:94-103. [DOI: 10.1124/dmd.118.083733] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/27/2018] [Indexed: 12/14/2022] Open
|
29
|
Yoshikado T, Toshimoto K, Maeda K, Kusuhara H, Kimoto E, Rodrigues AD, Chiba K, Sugiyama Y. PBPK Modeling of Coproporphyrin I as an Endogenous Biomarker for Drug Interactions Involving Inhibition of Hepatic OATP1B1 and OATP1B3. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2018; 7:739-747. [PMID: 30175555 PMCID: PMC6263667 DOI: 10.1002/psp4.12348] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 07/06/2018] [Indexed: 12/22/2022]
Abstract
The aim of the present study was to establish a physiologically based pharmacokinetic (PBPK) model for coproporphyrin I (CP-I), a biomarker supporting the prediction of drug-drug interactions (DDIs) involving hepatic organic anion transporting polypeptide 1B (OATP1B), using clinical DDI data with an OATP1B inhibitor rifampicin (300 and 600 mg, orally). The in vivo inhibition constants of rifampicin used as initial input parameters for OATP1Bs (Ki,u,OATP1Bs ) and multidrug resistance-associated protein two-mediated biliary excretion were estimated as 0.23 and 0.87 μM, respectively, from previous reports. Sensitivity analysis demonstrated that the Ki,u,OATP1Bs and biosynthesis rate of CP-I affected the magnitude of the interaction. Ki,u,OATP1Bs values optimized by nonlinear least-squares fitting were ~0.5-fold of the initial value. It was determined that the blood concentration-time profiles of four statins were well-predicted using corrected individual Ki,u,OATP1B values (ratio of in vitro Ki,u(statin) /in vitro Ki,u(CP-I) ). In conclusion, PBPK modeling of CP-I supports dynamic prediction of OATP1B-mediated DDIs.
Collapse
Affiliation(s)
- Takashi Yoshikado
- Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Yokohama, Kanagawa, Japan.,Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Yokohama, Kanagawa, Japan
| | - Kota Toshimoto
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Yokohama, Kanagawa, Japan
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Emi Kimoto
- Transporter Sciences Group, ADME Sciences, Medicine Design, Pfizer, Groton, Connecticut, USA
| | - A David Rodrigues
- Transporter Sciences Group, ADME Sciences, Medicine Design, Pfizer, Groton, Connecticut, USA
| | - Koji Chiba
- Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Yokohama, Kanagawa, Japan
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Yokohama, Kanagawa, Japan
| |
Collapse
|
30
|
Ishida T, Tokuda K, Hisaka A, Honma M, Kijima S, Takatoku H, Iwatsubo T, Moritoyo T, Suzuki H. A Novel Method to Estimate Long-Term Chronological Changes From Fragmented Observations in Disease Progression. Clin Pharmacol Ther 2018; 105:436-447. [PMID: 29951994 PMCID: PMC6617977 DOI: 10.1002/cpt.1166] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/14/2018] [Indexed: 12/16/2022]
Abstract
Clinical observations of patients with chronic diseases are often restricted in terms of duration. Therefore, obtaining a quantitative and comprehensive understanding of the chronology of chronic diseases is challenging, because of the inability to precisely estimate the patient's disease stage at the time point of observation. We developed a novel method to reconstitute long-term disease progression from temporally fragmented data by extending the nonlinear mixed-effects model to incorporate the estimation of "disease time" of each subject. Application of this method to sporadic Alzheimer's disease successfully depicted disease progression over 20 years. The covariate analysis revealed earlier onset of amyloid-β accumulation in male and female apolipoprotein E ε4 homozygotes, whereas disease progression was remarkably slower in female ε3 homozygotes compared with female ε4 carriers and males. Simulation of a clinical trial suggests patient recruitment using the information of precise disease time of each patient will decrease the sample size required for clinical trials.
Collapse
Affiliation(s)
- Takaaki Ishida
- Department of Pharmacy, Faculty of Medicine, The University of Tokyo Hospital, The University of Tokyo, Tokyo, Japan
| | - Keita Tokuda
- Department of Pharmacy, Faculty of Medicine, The University of Tokyo Hospital, The University of Tokyo, Tokyo, Japan
| | - Akihiro Hisaka
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Masashi Honma
- Department of Pharmacy, Faculty of Medicine, The University of Tokyo Hospital, The University of Tokyo, Tokyo, Japan
| | - Shinichi Kijima
- Advanced Review With Electronic Data Promotion Group, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Hiroyuki Takatoku
- Office of New Drug II, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takashi Moritoyo
- Office of New Drug II, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Department of Clinical Research Governance, Faculty of Medicine, The University of Tokyo Hospital, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Suzuki
- Department of Pharmacy, Faculty of Medicine, The University of Tokyo Hospital, The University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
31
|
Suzuki C, Uehara T, Kanazawa N, Wada S, Suzuki H, Arano Y. Preferential Cleavage of a Tripeptide Linkage by Enzymes on Renal Brush Border Membrane To Reduce Renal Radioactivity Levels of Radiolabeled Antibody Fragments. J Med Chem 2018; 61:5257-5268. [DOI: 10.1021/acs.jmedchem.8b00198] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Chie Suzuki
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana,
Chuo-ku, Chiba 260-8675, Japan
- Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Tomoya Uehara
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana,
Chuo-ku, Chiba 260-8675, Japan
| | - Naoki Kanazawa
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana,
Chuo-ku, Chiba 260-8675, Japan
| | - Shota Wada
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana,
Chuo-ku, Chiba 260-8675, Japan
| | - Hiroyuki Suzuki
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana,
Chuo-ku, Chiba 260-8675, Japan
| | - Yasushi Arano
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana,
Chuo-ku, Chiba 260-8675, Japan
| |
Collapse
|
32
|
Yao Y, Toshimoto K, Kim SJ, Yoshikado T, Sugiyama Y. Quantitative Analysis of Complex Drug-Drug Interactions between Cerivastatin and Metabolism/Transport Inhibitors Using Physiologically Based Pharmacokinetic Modeling. Drug Metab Dispos 2018; 46:924-933. [PMID: 29712725 DOI: 10.1124/dmd.117.079210] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 04/25/2018] [Indexed: 02/06/2023] Open
Abstract
Cerivastatin (CER) was withdrawn from the world market because of lethal rhabdomyolysis. Coadministrations of CER and cyclosporine A (CsA) or gemfibrozil (GEM) have been reported to increase the CER blood concentration. CsA is an inhibitor of organic anion transporting polypeptide (OATP)1B1 and CYP3A4, and GEM and its glucuronide (GEM-glu) inhibit OATP1B1 and CYP2C8. The purpose of this study was to describe the transporter-/enzyme-mediated drug-drug interactions (DDIs) of CER with CsA or GEM based on unified physiologically based pharmacokinetic (PBPK) models and to investigate whether the DDIs can be quantitatively analyzed by a bottom-up approach. Initially, the PBPK models for CER and GEM/GEM-glu were constructed based on the previously reported standard protocols. Next, the drug-dependent parameters were optimized by Cluster Newton Method. Thus, described concentration-time profiles for CER and GEM/GEM-glu agreed well with the clinically observed data. The DDIs were then simulated using the established PBPK models with previously obtained in vitro inhibition constants of CsA or GEM/GEM-glu against the OATP1B1 and cytochrome P450s. DDIs with the inhibitors were underestimated compared with observed data using the geometric means of reported values. To search for better described parameters within the range of in vitro values, sensitivity analyses were performed for DDIs of CER. Using the in vitro parameter sets selected by sensitivity analyses, these DDIs were well reproduced, indicating that the present PBPK models were able to describe adequately the clinical DDIs based on a bottom-up approach. The approaches in this study would be applicable to the prediction of other DDIs involving both transporters and metabolic enzymes.
Collapse
Affiliation(s)
- Yoshiaki Yao
- Analysis & Pharmacokinetics Research Laboratories, Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan (Y.Y.), and Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Kanagawa, Japan (K.T., S.K., T.Y., Y.S.)
| | - Kota Toshimoto
- Analysis & Pharmacokinetics Research Laboratories, Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan (Y.Y.), and Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Kanagawa, Japan (K.T., S.K., T.Y., Y.S.)
| | - Soo-Jin Kim
- Analysis & Pharmacokinetics Research Laboratories, Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan (Y.Y.), and Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Kanagawa, Japan (K.T., S.K., T.Y., Y.S.)
| | - Takashi Yoshikado
- Analysis & Pharmacokinetics Research Laboratories, Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan (Y.Y.), and Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Kanagawa, Japan (K.T., S.K., T.Y., Y.S.)
| | - Yuichi Sugiyama
- Analysis & Pharmacokinetics Research Laboratories, Drug Discovery Research, Astellas Pharma Inc., Ibaraki, Japan (Y.Y.), and Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Kanagawa, Japan (K.T., S.K., T.Y., Y.S.)
| |
Collapse
|
33
|
Ochiai W, Kobayashi H, Kitaoka S, Kashiwada M, Koyama Y, Nakaishi S, Nagai T, Aburada M, Sugiyama K. Effect of the active ingredient of Kaempferia parviflora, 5,7-dimethoxyflavone, on the pharmacokinetics of midazolam. J Nat Med 2018; 72:607-614. [PMID: 29550915 DOI: 10.1007/s11418-018-1184-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/26/2018] [Indexed: 11/28/2022]
Abstract
5,7-Dimethoxyflavone (5,7-DMF), one of the major components of Kaempferia parviflora, has anti-obesity, anti-inflammatory, and antineoplastic effects. On the other hand, in vitro studies have reported that it directly inhibits the drug metabolizing enzyme family cytochrome P450 (CYP) 3As. In this study, its safety was evaluated from a pharmacokinetic point of view, based on daily ingestion of 5,7-DMF. Midazolam, a substrate of CYP3As, was orally administered to mice treated with 5,7-DMF for 10 days, and its pharmacokinetic properties were investigated. In the group administered 5,7-DMF, the area under the curve (AUC) of midazolam increased by 130% and its biological half-life was extended by approximately 100 min compared to the control group. Compared to the control group, 5,7-DMF markedly decreased the expression of CYP3A11 and CYP3A25 in the liver. These results suggest that continued ingestion of 5,7-DMF decreases the expression of CYP3As in the liver, consequently increasing the blood concentrations of drugs metabolized by CYP3As.
Collapse
Affiliation(s)
- Wataru Ochiai
- Department of Clinical Pharmacokinetics, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan.
| | - Hiroko Kobayashi
- Department of Clinical Pharmacokinetics, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Satoshi Kitaoka
- Department of Clinical Pharmacokinetics, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Mayumi Kashiwada
- Department of Clinical Pharmacokinetics, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Yuya Koyama
- Department of Clinical Pharmacokinetics, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Saho Nakaishi
- Department of Clinical Pharmacokinetics, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Tomomi Nagai
- Department of Clinical Pharmacokinetics, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Masaki Aburada
- Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585, Japan
| | - Kiyoshi Sugiyama
- Department of Functional Molecule, Kinetics Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan.
| |
Collapse
|
34
|
Asaumi R, Toshimoto K, Tobe Y, Hashizume K, Nunoya K, Imawaka H, Lee W, Sugiyama Y. Comprehensive PBPK Model of Rifampicin for Quantitative Prediction of Complex Drug-Drug Interactions: CYP3A/2C9 Induction and OATP Inhibition Effects. CPT Pharmacometrics Syst Pharmacol 2018; 7:186-196. [PMID: 29368402 PMCID: PMC5869557 DOI: 10.1002/psp4.12275] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/27/2017] [Accepted: 12/28/2017] [Indexed: 01/13/2023] Open
Abstract
This study aimed to construct a physiologically based pharmacokinetic (PBPK) model of rifampicin that can accurately and quantitatively predict complex drug-drug interactions (DDIs) involving its saturable hepatic uptake and auto-induction. Using in silico and in vitro parameters, and reported clinical pharmacokinetic data, rifampicin PBPK model was built and relevant parameters for saturable hepatic uptake and UDP-glucuronosyltransferase (UGT) auto-induction were optimized by fitting. The parameters for cytochrome P450 (CYP) 3A and CYP2C9 induction by rifampicin were similarly optimized using clinical DDI data with midazolam and tolbutamide as probe substrates, respectively. For validation, our current PBPK model was applied to simulate complex DDIs with glibenclamide (a substrate of CYP3A/2C9 and hepatic organic anion transporting polypeptides (OATPs)). Simulated results were in quite good accordance with the observed data. Altogether, our constructed PBPK model of rifampicin demonstrates the robustness and utility in quantitatively predicting CYP3A/2C9 induction-mediated and/or OATP inhibition-mediated DDIs with victim drugs.
Collapse
Affiliation(s)
- Ryuta Asaumi
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd.TsukubaIbarakiJapan
| | - Kota Toshimoto
- Sugiyama LaboratoryRIKEN Innovation Center, RIKENYokohamaKanagawaJapan
| | - Yoshifusa Tobe
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd.TsukubaIbarakiJapan
| | - Kenta Hashizume
- Drug Development Solutions Division, Sekisui Medical Co., Ltd.IbarakiJapan
| | - Ken‐ichi Nunoya
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd.TsukubaIbarakiJapan
| | - Haruo Imawaka
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd.TsukubaIbarakiJapan
| | - Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical SciencesSeoul National UniversitySeoulKorea
| | - Yuichi Sugiyama
- Sugiyama LaboratoryRIKEN Innovation Center, RIKENYokohamaKanagawaJapan
| |
Collapse
|
35
|
Sato M, Toshimoto K, Tomaru A, Yoshikado T, Tanaka Y, Hisaka A, Lee W, Sugiyama Y. Physiologically Based Pharmacokinetic Modeling of Bosentan Identifies the Saturable Hepatic Uptake As a Major Contributor to Its Nonlinear Pharmacokinetics. Drug Metab Dispos 2018; 46:740-748. [PMID: 29475833 DOI: 10.1124/dmd.117.078972] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 02/21/2018] [Indexed: 01/02/2023] Open
Abstract
Bosentan is a substrate of hepatic uptake transporter organic anion-transporting polypeptides (OATPs), and undergoes extensive hepatic metabolism by cytochrome P450 (P450), namely, CYP3A4 and CYP2C9. Several clinical investigations have reported a nonlinear relationship between bosentan doses and its systemic exposure, which likely involves the saturation of OATP-mediated uptake, P450-mediated metabolism, or both in the liver. Yet, the underlying causes for the nonlinear bosentan pharmacokinetics are not fully delineated. To address this, we performed physiologically based pharmacokinetic (PBPK) modeling analyses for bosentan after its intravenous administration at different doses. As a bottom-up approach, PBPK modeling analyses were performed using in vitro kinetic parameters, other relevant parameters, and scaling factors. As top-down approaches, three different types of PBPK models that incorporate the saturation of hepatic uptake, metabolism, or both were compared. The prediction from the bottom-up approach (models 1 and 2) yielded blood bosentan concentration-time profiles and their systemic clearance values that were not in good agreement with the clinically observed data. From top-down approaches (models 3, 4, 5-1, and 5-2), the prediction accuracy was best only with the incorporation of the saturable hepatic uptake for bosentan. Taken together, the PBPK models for bosentan were successfully established, and the comparison of different PBPK models identified the saturation of the hepatic uptake process as a major contributing factor for the nonlinear pharmacokinetics of bosentan.
Collapse
Affiliation(s)
- Masanobu Sato
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (M.S.); Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (K.T., A.T., T.Y., Y.S.); DMPK Research Laboratory, Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., Tochigi, Japan (Y.T); Graduate School and Faculty of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.H.); and College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea (W.L.)
| | - Kota Toshimoto
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (M.S.); Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (K.T., A.T., T.Y., Y.S.); DMPK Research Laboratory, Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., Tochigi, Japan (Y.T); Graduate School and Faculty of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.H.); and College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea (W.L.)
| | - Atsuko Tomaru
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (M.S.); Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (K.T., A.T., T.Y., Y.S.); DMPK Research Laboratory, Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., Tochigi, Japan (Y.T); Graduate School and Faculty of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.H.); and College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea (W.L.)
| | - Takashi Yoshikado
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (M.S.); Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (K.T., A.T., T.Y., Y.S.); DMPK Research Laboratory, Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., Tochigi, Japan (Y.T); Graduate School and Faculty of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.H.); and College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea (W.L.)
| | - Yuta Tanaka
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (M.S.); Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (K.T., A.T., T.Y., Y.S.); DMPK Research Laboratory, Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., Tochigi, Japan (Y.T); Graduate School and Faculty of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.H.); and College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea (W.L.)
| | - Akihiro Hisaka
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (M.S.); Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (K.T., A.T., T.Y., Y.S.); DMPK Research Laboratory, Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., Tochigi, Japan (Y.T); Graduate School and Faculty of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.H.); and College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea (W.L.)
| | - Wooin Lee
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (M.S.); Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (K.T., A.T., T.Y., Y.S.); DMPK Research Laboratory, Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., Tochigi, Japan (Y.T); Graduate School and Faculty of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.H.); and College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea (W.L.)
| | - Yuichi Sugiyama
- Advanced Review with Electronic Data Promotion Group, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan (M.S.); Sugiyama Laboratory, RIKEN Innovation Center, Research Cluster for Innovation, RIKEN, Kanagawa, Japan (K.T., A.T., T.Y., Y.S.); DMPK Research Laboratory, Watarase Research Center, Kyorin Pharmaceutical Co., Ltd., Tochigi, Japan (Y.T); Graduate School and Faculty of Pharmaceutical Sciences, Chiba University, Chiba, Japan (A.H.); and College of Pharmacy, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea (W.L.)
| |
Collapse
|
36
|
Futatsugi A, Toshimoto K, Yoshikado T, Sugiyama Y, Kato Y. Evaluation of Alteration in Hepatic and Intestinal BCRP Function In Vivo from ABCG2 c.421C>A Polymorphism Based on PBPK Analysis of Rosuvastatin. Drug Metab Dispos 2018; 46:749-757. [PMID: 29440178 DOI: 10.1124/dmd.117.078816] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 02/08/2018] [Indexed: 01/06/2023] Open
Abstract
Polymorphism c.421C>A in the ABCG2 gene is thought to reduce the activity of breast cancer resistance protein (BCRP), a xenobiotic transporter, although it is not clear which organ(s) contributes to the polymorphism-associated pharmacokinetic change. The aim of the present study was to estimate quantitatively the influence of c.421C>A on intestinal and hepatic BCRP activity using a physiologically based pharmacokinetic (PBPK) model of rosuvastatin developed from clinical data and several in vitro studies. Simultaneous fitting of clinical data for orally and intravenously administered rosuvastatin, obtained in human subjects without genotype information, was first performed with the PBPK model to estimate intrinsic clearance for hepatic elementary process. The fraction of BCRP activity in 421CA and 421AA (fca and faa values, respectively) with respect to that in 421CC subjects was then estimated based on extended clearance concepts and simultaneous fitting to oral administration data for the three genotypes (421CC, 421CA, and 421AA). On the assumption that c.421C>A affects both intestinal and hepatic BCRP, clinical data in each genotype were well reproduced by the model, and the estimated terminal half-life was compatible with the observed values. The assumption that c.421C>A affects only either intestinal or hepatic BCRP gave poorer agreement with observed values. The faa values obtained on the former assumption were 0.48-0.54. Thus, PBPK model analysis enabled quantitative evaluation of alteration in BCRP activity owing to c.421C>A, and BCRP activity in 421AA was estimated as half that in 421CC.
Collapse
Affiliation(s)
- Azusa Futatsugi
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Cluster for Industry Partnerships, RIKEN (A.F., K.T., T.Y., Y.S.), and Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University (A.F., Y.K.), Kanazawa, Japan
| | - Kota Toshimoto
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Cluster for Industry Partnerships, RIKEN (A.F., K.T., T.Y., Y.S.), and Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University (A.F., Y.K.), Kanazawa, Japan
| | - Takashi Yoshikado
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Cluster for Industry Partnerships, RIKEN (A.F., K.T., T.Y., Y.S.), and Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University (A.F., Y.K.), Kanazawa, Japan
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Cluster for Industry Partnerships, RIKEN (A.F., K.T., T.Y., Y.S.), and Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University (A.F., Y.K.), Kanazawa, Japan
| | - Yukio Kato
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Cluster for Industry Partnerships, RIKEN (A.F., K.T., T.Y., Y.S.), and Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University (A.F., Y.K.), Kanazawa, Japan
| |
Collapse
|
37
|
Kim SJ, Toshimoto K, Yao Y, Yoshikado T, Sugiyama Y. Quantitative Analysis of Complex Drug–Drug Interactions Between Repaglinide and Cyclosporin A/Gemfibrozil Using Physiologically Based Pharmacokinetic Models With In Vitro Transporter/Enzyme Inhibition Data. J Pharm Sci 2017; 106:2715-2726. [DOI: 10.1016/j.xphs.2017.04.063] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/17/2017] [Accepted: 04/24/2017] [Indexed: 12/14/2022]
|
38
|
Quantitative Analyses of the Influence of Parameters Governing Rate-Determining Process of Hepatic Elimination of Drugs on the Magnitudes of Drug-Drug Interactions via Hepatic OATPs and CYP3A Using Physiologically Based Pharmacokinetic Models. J Pharm Sci 2017; 106:2739-2750. [DOI: 10.1016/j.xphs.2017.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/20/2017] [Accepted: 05/01/2017] [Indexed: 01/20/2023]
|
39
|
Yamamoto T, Furihata K, Hisaka A, Moritoyo T, Ogoe K, Kusayama S, Motohashi K, Mori A, Iwatsubo T, Suzuki H. Notable Drug-Drug Interaction Between Etizolam and Itraconazole in Poor Metabolizers of Cytochrome P450 2C19. J Clin Pharmacol 2017; 57:1491-1499. [PMID: 28679023 DOI: 10.1002/jcph.956] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 05/08/2017] [Indexed: 01/16/2023]
Abstract
In this study, impact of a polymorphism of CYP2C19 on drug-drug interaction (DDI) was examined for etizolam. The effect of itraconazole (a strong CYP3A inhibitor) on the pharmacokinetics of etizolam (a substrate of CYP2C19 and CYP3A) was assessed in both extensive metabolizers (EMs) and poor metabolizers (PMs) of CYP2C19. Sixteen participants (8 EMs and 8 PMs) received a single oral dose of etizolam (0.25 mg) on day 1. The participants ingested itraconazole (200 mg twice a day) on days 2-5. On day 5, participants received an oral dose of etizolam (0.25 mg) again. Before coadministration of itraconazole (day 1), the area under the time-plasma concentration curve from time zero to infinity (AUC∞ ) of etizolam was higher in PMs than in EMs (2.65-fold, P < .01). Coadministration of itraconazole increased the AUC∞ of etizolam 1.66-fold and 2.34-fold in EMs and PMs, respectively (day 5). Consequently, AUC∞ was 6.18-fold higher in PMs with itraconazole than that in EMs without itraconazole. The increase by itraconazole was larger in PMs (P < .01). In heterozygous EMs (hEMs), AUC∞ was simulated to be 2.56-fold higher with itraconazole than that in EMs without itraconazole. We found that in vitro measurements of fraction metabolized (fm ) using the liver microsome prepared from PM donors would be helpful to predict polymorphism-dependent DDIs. These results suggest that the PMs and hEMs of a polymorphic CYP would be at higher risk of DDIs relative to EMs for drugs metabolized by both polymorphic and nonpolymorphic CYPs such as etizolam.
Collapse
Affiliation(s)
- Takehito Yamamoto
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | - Kenichi Furihata
- P-One Clinic, Keikokai Medical Corporation, Tokyo, Japan.,Department of Clinical Pharmacology, Tokai University School of Medicine, Kanagawa, Japan
| | - Akihiro Hisaka
- Pharmacology and Pharmacokinetics, The University of Tokyo Hospital.,Present affiliation: Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Takashi Moritoyo
- Department of Clinical Research Governance, The University of Tokyo Hospital, Tokyo, Japan
| | - Kazuaki Ogoe
- P-One Clinic, Keikokai Medical Corporation, Tokyo, Japan
| | | | - Keiju Motohashi
- Unit for Early and Exploratory Clinical Department, The University of Tokyo Hospital, Tokyo, Japan
| | - Akiko Mori
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | - Takeshi Iwatsubo
- Unit for Early and Exploratory Clinical Department, The University of Tokyo Hospital, Tokyo, Japan.,Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Suzuki
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
40
|
Fukuchi Y, Toshimoto K, Mori T, Kakimoto K, Tobe Y, Sawada T, Asaumi R, Iwata T, Hashimoto Y, Nunoya KI, Imawaka H, Miyauchi S, Sugiyam Y. Analysis of Nonlinear Pharmacokinetics of a Highly Albumin-Bound Compound: Contribution of Albumin-Mediated Hepatic Uptake Mechanism. J Pharm Sci 2017; 106:2704-2714. [PMID: 28465151 DOI: 10.1016/j.xphs.2017.04.052] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/20/2017] [Accepted: 04/20/2017] [Indexed: 12/20/2022]
Abstract
The cause of nonlinear pharmacokinetics (PK) (more than dose-proportional increase in exposure) of a urea derivative under development (compound A: anionic compound [pKa: 4.4]; LogP: 6.5; and plasma protein binding: 99.95%) observed in a clinical trial was investigated. Compound A was metabolized by CYP3A4, UGT1A1, and UGT1A3 with unbound Km of 3.3-17.8 μmol/L. OATP1B3-mediated uptake of compound A determined in the presence of human serum albumin (HSA) showed that unbound Km and Vmax decreased with increased HSA concentration. A greater decrease in unbound Km than in Vmax resulted in increased uptake clearance (Vmax/unbound Km) with increased HSA concentration, the so-called albumin-mediated uptake. At 2% HSA concentration, unbound Km was 0.00657 μmol/L. A physiologically based PK model assuming saturable hepatic uptake nearly replicated clinical PK of compound A. Unbound Km for hepatic uptake estimated from the model was 0.000767 μmol/L, lower than the in vitro unbound Km at 2% HSA concentration, whereas decreased Km with increased concentration of HSA in vitro indicated lower Km at physiological HSA concentration (4%-5%). In addition, unbound Km values for metabolizing enzymes were much higher than unbound Km for OATP1B3, indicating that the nonlinear PK of compound A is primarily attributed to saturated OATP1B3-mediated hepatic uptake of compound A.
Collapse
Affiliation(s)
- Yukina Fukuchi
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Company, Ltd., Ibaraki, Japan
| | - Kota Toshimoto
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Kanagawa, Japan
| | - Takanori Mori
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Company, Ltd., Ibaraki, Japan
| | - Keisuke Kakimoto
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Company, Ltd., Ibaraki, Japan
| | - Yoshifusa Tobe
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Company, Ltd., Ibaraki, Japan
| | - Takeshi Sawada
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Company, Ltd., Ibaraki, Japan
| | - Ryuta Asaumi
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Company, Ltd., Ibaraki, Japan
| | - Takeyuki Iwata
- Oncology Clinical Development Planning, Ono Pharmaceutical Company, Ltd., Osaka, Japan
| | - Yoshitaka Hashimoto
- Translational Medicine Center, Ono Pharmaceutical Company, Ltd., Osaka, Japan
| | - Ken-Ichi Nunoya
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Company, Ltd., Ibaraki, Japan
| | - Haruo Imawaka
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Company, Ltd., Ibaraki, Japan.
| | - Seiji Miyauchi
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Kanagawa, Japan
| | - Yuichi Sugiyam
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Kanagawa, Japan
| |
Collapse
|
41
|
Matsui K, Tsume Y, Takeuchi S, Searls A, Amidon GL. Utilization of Gastrointestinal Simulator, an in Vivo Predictive Dissolution Methodology, Coupled with Computational Approach To Forecast Oral Absorption of Dipyridamole. Mol Pharm 2017; 14:1181-1189. [DOI: 10.1021/acs.molpharmaceut.6b01063] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Kazuki Matsui
- College
of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109-1065, United States
- Drug
Metabolism and Pharmacokinetics, Research Center, Mochida Pharmaceutical Company Limited, 722 Uenohara, Jimba, Gotemba, Shizuoka 412-8524, Japan
| | - Yasuhiro Tsume
- College
of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109-1065, United States
| | - Susumu Takeuchi
- Pharmacokinetics
Group, Sawai Pharmaceutical Company Limited, 5-2-30, Miyahara, Yodogawa-ku, Osaka 532-0003, Japan
| | - Amanda Searls
- College
of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109-1065, United States
| | - Gordon L. Amidon
- College
of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109-1065, United States
| |
Collapse
|
42
|
Estimation of the Contribution of CYP2C8 and CYP3A4 in Repaglinide Metabolism by Human Liver Microsomes Under Various Buffer Conditions. J Pharm Sci 2017; 106:2847-2852. [PMID: 28238899 DOI: 10.1016/j.xphs.2017.02.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 02/15/2017] [Accepted: 02/17/2017] [Indexed: 11/21/2022]
Abstract
We have previously reported that the microsomal activities of CYP2C8 and CYP3A4 largely depend on the buffer condition used in in vitro metabolic studies, with different patterns observed between the 2 isozymes. In the present study, therefore, the possibility of buffer condition dependence of the fraction metabolized by CYP2C8 (fm2C8) for repaglinide, a dual substrate of CYP2C8 and CYP3A4, was estimated using human liver microsomes under various buffer conditions. Montelukast and ketoconazole showed a potent and concentration-dependent inhibition of CYP2C8-mediated paclitaxel 6α-hydroxylation and CYP3A4-mediated triazolam α-hydroxylation, respectively, without dependence on the buffer condition. Repaglinide depletion was inhibited by both inhibitors, but the degree of inhibition depended on buffer conditions. Based on these results, the contribution of CYP2C8 in repaglinide metabolism was estimated to be larger than that of CYP3A4 under each buffer condition, and the fm2C8 value of 0.760, estimated in 50 mM phosphate buffer, was the closest to the value (0.801) estimated in our previous modeling analysis based on its concentration increase in a clinical drug interaction study. Researchers should be aware of the possibility of buffer condition affecting the estimated contribution of enzyme(s) in drug metabolism processes involving multiple enzymes.
Collapse
|
43
|
Kusuhara H, Maeda K. Determination of kinetic parameters for 123-I thyroid uptake in healthy Japanese. EPJ WEB OF CONFERENCES 2017. [DOI: 10.1051/epjconf/201715308007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
44
|
Yoshikado T, Yoshida K, Kotani N, Nakada T, Asaumi R, Toshimoto K, Maeda K, Kusuhara H, Sugiyama Y. Quantitative Analyses of Hepatic OATP-Mediated Interactions Between Statins and Inhibitors Using PBPK Modeling With a Parameter Optimization Method. Clin Pharmacol Ther 2016; 100:513-523. [PMID: 27170342 DOI: 10.1002/cpt.391] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/20/2016] [Accepted: 05/03/2016] [Indexed: 01/01/2023]
Abstract
This study aimed to construct a widely applicable method for quantitative analyses of drug-drug interactions (DDIs) caused by the inhibition of hepatic organic anion transporting polypeptides (OATPs) using physiologically based pharmacokinetic (PBPK) modeling. Models were constructed for pitavastatin, fluvastatin, and pravastatin as substrates and cyclosporin A (CsA) and rifampicin (RIF) as inhibitors, where enterohepatic circulations (EHC) of statins were incorporated. By fitting to clinical data, parameters that described absorption, hepatic elimination, and EHC processes were optimized, and the extent of these DDIs was explained satisfactorily. Similar in vivo inhibition constant (Ki ) values of each inhibitor against OATPs were obtained, regardless of the substrates. Estimated Ki values of CsA were comparable to reported in vitro values with the preincubation of CsA, while those of RIF were smaller than reported in vitro values (coincubation). In conclusion, this study proposes a method to optimize in vivo PBPK parameters in hepatic uptake transporter-mediated DDIs.
Collapse
Affiliation(s)
- T Yoshikado
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Yokohama, Kanagawa, Japan
| | - K Yoshida
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - N Kotani
- Clinical Pharmacology Strategy Group, Translational Clinical Research Science & Strategy Dept., Chugai Pharmaceutical Co., Tokyo, Japan
| | - T Nakada
- DMPK Research Laboratories Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma, Chiba, Japan
| | - R Asaumi
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ibaraki, Japan
| | - K Toshimoto
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Yokohama, Kanagawa, Japan
| | - K Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - H Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Y Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Yokohama, Kanagawa, Japan.
| |
Collapse
|
45
|
Shirasaka Y, Lee N, Zha W, Wagner D, Wang J. Involvement of organic cation transporter 3 (Oct3/Slc22a3) in the bioavailability and pharmacokinetics of antidiabetic metformin in mice. Drug Metab Pharmacokinet 2016; 31:385-388. [PMID: 27569291 DOI: 10.1016/j.dmpk.2016.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/06/2016] [Accepted: 04/21/2016] [Indexed: 01/11/2023]
Abstract
Metformin is widely used for the treatment of type II diabetes mellitus. It was reported to be substrate of OCT3/Oct3, which is expressed in the brush boarder membrane of the enterocytes. However, the role of OCT3/Oct3 in the intestinal absorption process of metformin remains obscure. In the present study, we aimed to clarify the impact of Oct3 on the oral bioavailability and pharmacokinetics of metformin in mice, by means of in vivo pharmacokinetic study using wild-type (Oct3+/+) and Oct3-knockout (Oct3-/-) mice. When metformin (8.0 mg/kg) was intravenously administered to male Oct3+/+ and Oct3-/- mice, AUC0-∞ of metformin was evaluated to be 659 ± 133 μg h/mL and 734 ± 213 μg h/mL, respectively. In the case of orally administered metformin (15 mg/kg), AUC0-∞ was 578 ± 158 μg h/mL and 449 ± 101 μg h/mL in Oct3+/+ and Oct3-/- mice, respectively. Based on these pharmacokinetic parameters, absolute bioavailability (F) of metformin in Oct3+/+ mice was evaluated as 46.8%, and it was significantly decreased to 32.6% in Oct3-/- mice. Taking into account the fact that metformin undergoes negligible metabolism, these results imply that intestinal absorption of metformin is mediated at least in part by Oct3 in mice.
Collapse
Affiliation(s)
- Yoshiyuki Shirasaka
- Department of Pharmaceutics, School of Pharmacy, University of Washington, H272 Health Sciences Building, Seattle, WA 98195-7610, USA; Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Nora Lee
- Department of Pharmaceutics, School of Pharmacy, University of Washington, H272 Health Sciences Building, Seattle, WA 98195-7610, USA
| | - Weibin Zha
- Department of Pharmaceutics, School of Pharmacy, University of Washington, H272 Health Sciences Building, Seattle, WA 98195-7610, USA
| | - David Wagner
- Department of Pharmaceutics, School of Pharmacy, University of Washington, H272 Health Sciences Building, Seattle, WA 98195-7610, USA
| | - Joanne Wang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, H272 Health Sciences Building, Seattle, WA 98195-7610, USA.
| |
Collapse
|
46
|
Tanaka Y, Kitamura Y, Maeda K, Sugiyama Y. Quantitative Analysis of the ABCG2 c.421C>A Polymorphism Effect on In Vivo Transport Activity of Breast Cancer Resistance Protein (BCRP) Using an Intestinal Absorption Model. J Pharm Sci 2015; 104:3039-48. [PMID: 25639366 DOI: 10.1002/jps.24366] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 12/08/2014] [Accepted: 01/05/2015] [Indexed: 12/26/2022]
Abstract
ABCG2 c.421C>A is one of the most frequent polymorphisms in ABCG2, which encodes the breast cancer resistance protein (BCRP). Clinical pharmacogenetic studies have shown that the plasma area under the concentration-time curve (AUC) values after oral administration of BCRP substrate drugs are significantly higher in subjects homozygous for the c.421C>A polymorphism (421AA) than in wild-type subjects (421CC). The aim of this study was to quantitatively estimate the in vivo decrease of BCRP function caused by the c.421C>A polymorphism based on clinical pharmacokinetic data. Assuming that the pharmacokinetic alteration is accounted for by intestinal BCRP, the ratio of the transport activity of the mutated BCRP to that of the wild-type was optimized by comparing calculations from an intestinal absorption model and clinical pharmacokinetic data. In conclusion, the in vivo intestinal BCRP transport activity in 421AA subjects is estimated to be approximately 23% of that in the 421CC subjects.
Collapse
Affiliation(s)
- Yuta Tanaka
- Discovery Research Laboratories, Kyorin Pharmaceutical Company, Ltd, Tochigi, Japan
| | - Yoshiaki Kitamura
- Discovery Research Laboratories, Kyorin Pharmaceutical Company, Ltd, Tochigi, Japan
| | - Kazuya Maeda
- Department of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Research Cluster for Innovation, Yokohama, Japan
| |
Collapse
|
47
|
Ando H, Hisaka A, Suzuki H. A New Physiologically Based Pharmacokinetic Model for the Prediction of Gastrointestinal Drug Absorption: Translocation Model. Drug Metab Dispos 2015; 43:590-602. [DOI: 10.1124/dmd.114.060038] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
48
|
Kosaka K, Watanabe T, Susukida T, Aoki S, Sekine S, Kume T, Ito K. Key determinants of the circulatory exposure of organic anions: differences in hepatic uptake between multidrug resistance-associated protein 2 (Mrp2)-deficient rats and wild-type rats. Xenobiotica 2014; 45:556-62. [DOI: 10.3109/00498254.2014.997820] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
49
|
Shirota K, Kaneko M, Sasaki M, Minato K, Fujikata A, Ohta S, Hisaka A, Suzuki H. Analysis of the disposition of a novel p38 MAPK inhibitor, AKP-001, and its metabolites in rats with a simple physiologically based pharmacokinetic model. Drug Metab Dispos 2014; 43:217-26. [PMID: 25422274 DOI: 10.1124/dmd.114.060046] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
5-[(2-Chloro-6-fluorophenyl)acetylamino]-3-(4-fluorophenyl)-4-(4-pyrimidinyl)isoxazole (AKP-001) is a potent p38 mitogen-activated protein kinase inhibitor that is being developed to specifically target the intestines for the treatment of inflammatory bowel disease. According to the ante-drug concept, AKP-001 was designed to be metabolized to inactive forms via the first-pass metabolism to avoid undesirable systemic exposure. The purpose of this study is to investigate the pharmacokinetic characteristics of AKP-001 and its metabolites (M1 and M2) in rats, utilizing a simple physiologically based pharmacokinetic (PBPK) model. In vitro metabolic activity of AKP-001 in the S9 fraction of rat liver was examined, and plasma concentration-time profiles were developed following intravenous and/or oral administration of AKP-001 and its metabolites. AKP-001 was primarily metabolized to M1; however, M2 was not detected in liver S9 fractions. In accordance with this observation in vitro, M2 was detected in plasma after oral dosing of AKP-001 with a lag time of 1.5 hours, but not after intravenous dosing. To analyze pharmacokinetics in rats in vivo, a simple PBPK model was developed by simultaneous fitting of the plasma concentrations after treatment with AKP-001 and its metabolites. The observed plasma concentration-time profiles of AKP-001 and metabolites were described by the model adequately. Intestinal and systemic exposures of AKP-001 were simulated using the model to assess the relationship between pharmacokinetics and efficacy/safety. Model analysis suggested that oral bioavailability of intestine-targeting ante-drugs should be low to avoid systemic side effects. The pharmacokinetic properties of AKP-001 meet this criterion owing to extensive first-pass metabolism.
Collapse
Affiliation(s)
- Kazuhiko Shirota
- Pharmacokinetic Research Department, Developmental Research Center (K.S., M.K., M.S., K.M., and A.F.) and Synthetic Research Department, Medicinal Research Center (S.O.), ASKA Pharmaceutical Co. Ltd., Kanagawa, Japan; Pharmacology and Pharmacokinetics (A.H.) and Department of Pharmacy (H.S.), The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Makoto Kaneko
- Pharmacokinetic Research Department, Developmental Research Center (K.S., M.K., M.S., K.M., and A.F.) and Synthetic Research Department, Medicinal Research Center (S.O.), ASKA Pharmaceutical Co. Ltd., Kanagawa, Japan; Pharmacology and Pharmacokinetics (A.H.) and Department of Pharmacy (H.S.), The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Makoto Sasaki
- Pharmacokinetic Research Department, Developmental Research Center (K.S., M.K., M.S., K.M., and A.F.) and Synthetic Research Department, Medicinal Research Center (S.O.), ASKA Pharmaceutical Co. Ltd., Kanagawa, Japan; Pharmacology and Pharmacokinetics (A.H.) and Department of Pharmacy (H.S.), The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kouichi Minato
- Pharmacokinetic Research Department, Developmental Research Center (K.S., M.K., M.S., K.M., and A.F.) and Synthetic Research Department, Medicinal Research Center (S.O.), ASKA Pharmaceutical Co. Ltd., Kanagawa, Japan; Pharmacology and Pharmacokinetics (A.H.) and Department of Pharmacy (H.S.), The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akira Fujikata
- Pharmacokinetic Research Department, Developmental Research Center (K.S., M.K., M.S., K.M., and A.F.) and Synthetic Research Department, Medicinal Research Center (S.O.), ASKA Pharmaceutical Co. Ltd., Kanagawa, Japan; Pharmacology and Pharmacokinetics (A.H.) and Department of Pharmacy (H.S.), The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shuji Ohta
- Pharmacokinetic Research Department, Developmental Research Center (K.S., M.K., M.S., K.M., and A.F.) and Synthetic Research Department, Medicinal Research Center (S.O.), ASKA Pharmaceutical Co. Ltd., Kanagawa, Japan; Pharmacology and Pharmacokinetics (A.H.) and Department of Pharmacy (H.S.), The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akihiro Hisaka
- Pharmacokinetic Research Department, Developmental Research Center (K.S., M.K., M.S., K.M., and A.F.) and Synthetic Research Department, Medicinal Research Center (S.O.), ASKA Pharmaceutical Co. Ltd., Kanagawa, Japan; Pharmacology and Pharmacokinetics (A.H.) and Department of Pharmacy (H.S.), The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Suzuki
- Pharmacokinetic Research Department, Developmental Research Center (K.S., M.K., M.S., K.M., and A.F.) and Synthetic Research Department, Medicinal Research Center (S.O.), ASKA Pharmaceutical Co. Ltd., Kanagawa, Japan; Pharmacology and Pharmacokinetics (A.H.) and Department of Pharmacy (H.S.), The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
50
|
Sawamoto K, Huong TT, Sugimoto N, Mizutani Y, Sai Y, Miyamoto KI. Mechanisms of Lower Maintenance Dose of Tacrolimus in Obese Patients. Drug Metab Pharmacokinet 2014; 29:341-7. [DOI: 10.2133/dmpk.dmpk-13-rg-110] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|