1
|
Yadav P, Wairkar S. Tofacitinib in focus: Fascinating voyage from conventional formulations to novel delivery systems. Int J Pharm 2025; 671:125253. [PMID: 39842741 DOI: 10.1016/j.ijpharm.2025.125253] [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: 11/28/2024] [Revised: 01/11/2025] [Accepted: 01/19/2025] [Indexed: 01/24/2025]
Abstract
Tofacitinib, a Janus kinase (JAK) inhibitor, has emerged as a primary therapeutic agent for managing autoimmune diseases such as rheumatoid arthritis, psoriatic arthritis, dermatitis and ulcerative colitis. By inhibiting the phosphorylation of JAK enzymes, tofacitinib prevents their activation within the JAK-STAT signaling pathway, which is vital for inflammatory responses. However, the tofacitinib delivery presents significant challenges, including pH-dependent solubility, poor permeability and susceptibility to oral degradation. This review provides an in-depth analysis of current and emerging formulations to enhance the delivery and efficiency of tofacitinib. This review highlights the physicochemical, pharmacodynamic and pharmacokinetic properties of tofacitinib. Additionally, it discusses various strategies, including oral modified release formulations, topical/transdermal delivery utilizing lipid-based and polymeric systems, and parenteral delivery systems. Recent advancements in nanotechnology, such as liposomes, micelles, keratinocyte exosomes, proposomes, proglycosomes, transethosomes, squalenyl nanoparticles and lyotropic liquid crystalline nanoparticles, are explored as potential nanocarriers to existing delivery constraints. The development of advanced tofacitinib delivery systems can address the challenges in its delivery and improve therapeutic outcomes and patient compliance, paving the way for enhanced treatment strategies in autoimmune and inflammatory conditions.
Collapse
Affiliation(s)
- Priti Yadav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra 400056, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra 400056, India.
| |
Collapse
|
2
|
Choi HG, Park SY, Bae SH, Chang SY, Kim SH. Loganin Ameliorates Acute Kidney Injury and Restores Tofacitinib Metabolism in Rats: Implications for Renal Protection and Drug Interaction. Biomol Ther (Seoul) 2024; 32:601-610. [PMID: 39091013 PMCID: PMC11392661 DOI: 10.4062/biomolther.2024.008] [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: 01/10/2024] [Revised: 03/08/2024] [Accepted: 03/19/2024] [Indexed: 08/04/2024] Open
Abstract
Tofacitinib, a Janus kinase (JAK) inhibitor used to treat rheumatoid arthritis, is metabolized through hepatic cytochrome P450 (CYP), specifically CYP3A1/2 and CYP2C11. Prolonged administration of rheumatoid arthritis medications is generally associated with an increased risk of renal toxicity. Loganin (LGN), an iridoid glycoside, has hepatorenal regenerative properties. This study investigates the potential of LGN to mitigate acute kidney injury (AKI) and its effects on the pharmacokinetics of tofacitinib in rats with cisplatin-induced AKI. Both intravenous and oral administration of tofacitinib to AKI rats significantly increased the area under the plasma concentration-time curve from time 0 to infinity (AUC) compared with control (CON) rats, an increase attributed to the decelerated non-renal clearance (CLNR) and renal clearance (CLR) of tofacitinib. Administration of LGN to AKI rats, however, protected kidneys from severe impairment, restoring the pharmacokinetic parameters (AUC, CLNR, and CLR) of tofacitinib to those observed in untreated CON rats, with partial recovery of kidney function, as evidenced by an increase in creatinine clearance (CLCR). Possible interactions between drugs and natural components should be considered, especially when co-administering both a drug and a natural extract containing LGN or iridoid glycosides to patients with kidney injury.
Collapse
Affiliation(s)
- Hyeon Gyeom Choi
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - So Yeon Park
- Department of Biohealth Regulatory Science, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Sung Hun Bae
- AI-Superconvergence KIURI Translational Research Center, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Sun-Young Chang
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon 16499, Republic of Korea
- Department of Biohealth Regulatory Science, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - So Hee Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon 16499, Republic of Korea
- Department of Biohealth Regulatory Science, Graduate School of Ajou University, Suwon 16499, Republic of Korea
- AI-Superconvergence KIURI Translational Research Center, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| |
Collapse
|
3
|
Zou J, Jiang K, Chen Y, Ma Y, Xia C, Ding W, Yao M, Lin Y, Chen Y, Zhao Y, Gao F. Tofacitinib Citrate Coordination-Based Dual-Responsive/Scavenge Nanoplatform Toward Regulate Colonic Inflammatory Microenvironment for Relieving Colitis. Adv Healthc Mater 2024:e2401869. [PMID: 39180276 DOI: 10.1002/adhm.202401869] [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: 05/20/2024] [Revised: 07/16/2024] [Indexed: 08/26/2024]
Abstract
Ulcerative colitis is an inflammation of the colon characterized by immune dysregulation and intestinal inflammation. Developing safe oral nanomedicines that suppress intestinal inflammation, while modulating colonic inflammatory microenvironment by scavenging reactive oxygen species (ROS) and hydrogen sulfide (H2S) is crucial for the effective treatment of colitis. Here, the tofacitinib citrate and copper coordination-based nanoparticle (TF-Cu nanoparticle, T-C) to dual-scavenge ROS and H2S by coordination competition is synthesized. Moreover, the coordination of T-C using computer simulation is explored. To enhance the acid stability and inflammatory targeting of T-C, it is encapsulated with hyaluronic acid-modified chitosan, along with a calcium pectinate coating (T-C@HP). Owing to the dual pH/pectinase-responsive characteristics of T-C@HP, the nanoplatform can target inflamed colonic lesions, inhibiting phosphorylated Janus kinase 1. Furthermore, T-C@HP scavenges ROS and H2S, as well as increases NADPH levels, which is investigated by combining biosensor (HyPer7 and iNap1/c) and chemical probes. T-C@HP also alleviates colitis by regulating the colonic inflammatory microenvironment through multiple processes, including the modulation of apoptosis, macrophage polarization, tight junction, mucus layer, and intestinal flora. Complemented by satisfactory anti-inflammatory and biosafety results, this nanoplatform represents a promising, effective, and safe treatment option for colitis patients.
Collapse
Affiliation(s)
- Jiafeng Zou
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Kun Jiang
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - You Chen
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Ying Ma
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Chuanhe Xia
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Wenxing Ding
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Min Yao
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yiting Lin
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yanzuo Chen
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yuzheng Zhao
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
- Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
- Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, 100050, China
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Feng Gao
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
- Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai, 200237, China
- Engineering Research Center of Pharmaceutical Process Chemistry, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| |
Collapse
|
4
|
Wang Y, Zhou Q, Wang H, Song W, Wang J, Mamun AA, Geng P, Zhou Y, Wang S. Effect of P. corylifolia on the pharmacokinetic profile of tofacitinib and the underlying mechanism. Front Pharmacol 2024; 15:1351882. [PMID: 38650629 PMCID: PMC11033359 DOI: 10.3389/fphar.2024.1351882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/19/2024] [Indexed: 04/25/2024] Open
Abstract
This work aimed to explore the mechanisms underlying the interaction of the active furanocoumarins in P. corylifolia on tofacitinib both in vivo and in vitro. The concentration of tofacitinib and its metabolite M8 was determined using UPLC-MS/MS. The peak area ratio of M8 to tofacitinib was calculated to compare the inhibitory ability of furanocoumarin contained in the traditional Chinese medicine P. corylifolia in rat liver microsomes (RLMs), human liver microsomes (HLMs) and recombinant human CYP3A4 (rCYP3A4). We found that bergapten and isopsoralen exhibited more significant inhibitory activity in RLMs than other furanocoumarins. Bergapten and isopsoralen were selected to investigate tofacitinib drug interactions in vitro and in vivo. Thirty rats were randomly allocated into 5 groups (n = 6): control (0.5% CMC-Na), low-dose bergapten (20 mg/kg), high-dose bergapten (50 mg/kg), low-dose isopsoralen (20 mg/kg) and ketoconazole. 10 mg/kg of tofacitinib was orally intervented to each rat and the concentration level of tofacitinib in the rats were determined by UPLC-MS/MS. More imporrantly, the results showed that bergapten and isopsoralen significantly inhibited the metabolism of tofacitinib metabolism. The AUC(0-t), AUC(0-∞), MRT(0-t), MRT(0-∞) and Cmax of tofacitinib increased in varying degrees compared with the control group (all p < 0.05), but CLz/F decreased in varying degrees (p < 0.05) in the different dose bergapten group and isopsoralen group. Bergapten, isopsoralen and tofacitinib exhibit similar binding capacities with CYP3A4 by AutoDock 4.2 software, confirming that they compete for tofacitinib metabolism. P. corylifolia may considerably impact the metabolism of tofacitinib, which can provide essential information for the accurate therapeutic application of tofacitinib.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yunfang Zhou
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, Wenzhou Medical University Lishui Hospital, Lishui People’s Hospital, Lishui, Zhejiang, China
| | - Shuanghu Wang
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, Wenzhou Medical University Lishui Hospital, Lishui People’s Hospital, Lishui, Zhejiang, China
| |
Collapse
|
5
|
Shi Y, Lu Z, Song W, Wang Y, Zhou Q, Geng P, Zhou Y, Wang S, Han A. The Impact of Baohuoside I on the Metabolism of Tofacitinib in Rats. Drug Des Devel Ther 2024; 18:931-939. [PMID: 38560524 PMCID: PMC10980839 DOI: 10.2147/dddt.s436549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
Purpose To study the potential drug-drug interactions between tofacitinib and baohuoside I and to provide the scientific basis for rational use of them in clinical practice. Methods A total of eighteen Sprague-Dawley rats were randomly divided into three groups: control group, single-dose group (receiving a single dose of 20 mg/kg of baohuoside I), and multi-dose group (receiving multiple doses of baohuoside I for 7 days). On the seventh day, each rat was orally administered with 10 mg/kg of tofacitinib 30 minutes after giving baohuoside I or vehicle. Blood samples were collected and determined using UPLC-MS/MS. In vitro effects of baohuoside I on tofacitinib was investigated in rat liver microsomes (RLMs), as well as the underlying mechanism of inhibition. The semi-inhibitory concentration value (IC50) of baohuoside I was subsequently determined and its inhibitory mechanism against tofacitinib was analyzed. Furthermore, the interactions between baohuoside I, tofacitinib and CYP3A4 were explored using Pymol molecular docking simulation. Results The administration of baohuoside I orally has been observed to enhance the area under the concentration-time curve (AUC) of tofacitinib and decrease the clearance (CL). The observed disparity between the single-dose and multi-dose groups was statistically significant. Furthermore, our findings suggest that the impact of baohuoside I on tofacitinib metabolism may be a mixture of non-competitive and competitive inhibition. Baohuoside I exhibit an interaction with arginine (ARG) at position 106 of the CYP3A4 enzyme through hydrogen bonding, positioning itself closer to the site of action compared to tofacitinib. Conclusion Our study has demonstrated the presence of drug-drug interactions between baohuoside I and tofacitinib, which may arise upon pre-administration of tofacitinib. Altogether, our data indicated that an interaction existed between tofacitinib and baohuoside I and additional cares might be taken when they were co-administrated in clinic.
Collapse
Affiliation(s)
- Yaru Shi
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| | - Zebei Lu
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| | - Wei Song
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| | - Yu Wang
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| | - Quan Zhou
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| | - Peiwu Geng
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| | - Yunfang Zhou
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| | - Shuanghu Wang
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| | - Aixia Han
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, Zhejiang, 323000, People’s Republic of China
| |
Collapse
|
6
|
Bouali W, Kurtay G, Genç AA, Ahmed HEH, Soylak M, Erk N, Karimi-Maleh H. Nanodiamond (ND)-Based ND@CuAl 2O 4@Fe 3O 4 electrochemical sensor for Tofacitinib detection: A unified approach to integrate experimental data with DFT and molecular docking. ENVIRONMENTAL RESEARCH 2023; 238:117166. [PMID: 37741570 DOI: 10.1016/j.envres.2023.117166] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/23/2023] [Accepted: 09/15/2023] [Indexed: 09/25/2023]
Abstract
Tofacitinib (TOF) is gaining recognition as a potent therapeutic agent for a variety of autoimmune disorders, including rheumatoid arthritis and psoriasis. Ensuring precise drug concentration control during treatment necessitates a rapid and sensitive detection method. This study introduces a novel electrochemical sensor employing a composite of nanodiamond (ND), copper aluminate spinel oxide (CuAl2O4), and iron (II, III) oxide (Fe3O4) as modified materials for efficient TOF detection. Extensive analyses using physicochemical and electrochemical techniques were carried out to characterize the morphological, structural, and electrochemical properties of the ND@CuAl2O4@Fe3O4 composite. Thereafter, various voltammetric methods were utilized to evaluate the electrochemical behavior of the ND@CuAl2O4@Fe3O4-modified glassy carbon electrode (GCE) concerning TOF determination. The fabricated electrode showcased superior performance in electrochemical TOF detection in a buffered solution (pH = 5), achieving a remarkably low detection limit of 7.8 nM and a linear response from 0.05 μM to 13.21 μM. Furthermore, applying the modified electrode as an electrochemical sensor exhibited exceptional selectivity, stability, and practicality in determining TOF in pharmaceutical and biological samples. Alongside the sensor development, this study conducted a thorough investigation using Density Functional Theory (DFT) for the geometry optimization of TOF and the TOF-ND complex. Consequently performed molecular docking studies using Janus Kinase 1 (JAK1) (PDB ID: 3EYG) and JAK3 (PDB ID: 3LXK) indicated higher interaction of the TOF-ND conjugate with the JAKs, reflected by binding energies of -12.9 kcal/mol and -11.7 kcal/mol for JAK1 and JAK3 respectively, compared to -7.0 kcal/mol and -6.9 kcal/mol for TOF alone. These findings illustrate the potential of the ND-based ND@CuAl2O4@Fe3O4 composite as a proficient sensing material for TOF detection and the merits of DFT in providing a detailed understanding of the interactions at play. This pioneering research holds promise for real-time TOF monitoring, which will advance personalized treatment strategies and improve therapeutic outcomes for patients with autoimmune disorders.
Collapse
Affiliation(s)
- Wiem Bouali
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, 06560, Ankara, Turkey
| | - Gülbin Kurtay
- Hacettepe University, Faculty of Sciences, Department of Chemistry, 06800, Ankara, Turkey.
| | - Asena Ayşe Genç
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, 06560, Ankara, Turkey
| | | | - Mustafa Soylak
- Technology Research & Application Center (TAUM), Erciyes University, 38039, Kayseri, Turkey; Turkish Academy of Sciences (TUBA), Çankaya, Ankara, Turkey
| | - Nevin Erk
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, 06560, Ankara, Turkey.
| | - Hassan Karimi-Maleh
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China; School of Resources and Environment, University of Electronic Science and Technology of China, P.O. Box 611731, Xiyuan Ave, Chengdu, PR China; School of Engineering, Lebanese American University, Byblos, Lebanon
| |
Collapse
|
7
|
Won JM, Choi HG, Park SY, Kim JH, Kim SH. Effects of Hyperlipidemia on the Pharmacokinetics of Tofacitinib, a JAK 1/3 Inhibitor, in Rats. Pharmaceutics 2023; 15:2195. [PMID: 37765165 PMCID: PMC10534486 DOI: 10.3390/pharmaceutics15092195] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/08/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Tofacitinib, an inhibitor of Janus kinases (JAKs) 1 and 3, has been shown to be effective in the treatment of rheumatoid arthritis. The incidence of hyperlipidemia has been found to be higher in patients with rheumatoid arthritis. The present study therefore investigated the pharmacokinetics of tofacitinib after its intravenous (10 mg/kg) or oral (20 mg/kg) administration in poloxamer-407-induced hyperlipidemic (PHL) rats. The area under the plasma concentration-time curve from zero to infinity (AUC0-∞) after intravenous administration of tofacitinib was 73.5% higher in PHL than in control rats, owing to slower time-averaged nonrenal clearance (CLNR) in the former. Evaluation of in vitro metabolism showed that the intrinsic clearance (CLint) of tofacitinib was 38.6% lower in PHL than in control rats, owing to the decreased protein expression of hepatic cytochrome P450 (CYP) 3A1/2 and CYP2C11 in PHL rats. Similar results were observed in PHL rats after oral administration of tofacitinib. These results were likely due to the decreased CLNR, CLint, and P-glycoprotein (P-gp) expression in the intestines of PHL compared to control rats. Overall, these findings indicated that hyperlipidemia slowed the metabolism of tofacitinib, increasing its plasma concentrations, and that this reduced metabolism was due to alterations in expression of the proteins CYP3A1/2, CYP2C11, and P-gp in the liver and/or intestines of PHL rats.
Collapse
Affiliation(s)
- Jong Mun Won
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Hyeon Gyeom Choi
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - So Yeon Park
- Department of Biohealth Regulatory Science, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Jang-Hee Kim
- Department of Pathology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - So Hee Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon 16499, Republic of Korea
- Department of Biohealth Regulatory Science, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| |
Collapse
|
8
|
Bae SH, Choi HG, Park SY, Chang SY, Kim H, Kim SH. Effects of Isosakuranetin on Pharmacokinetic Changes of Tofacitinib in Rats with N-Dimethylnitrosamine-Induced Liver Cirrhosis. Pharmaceutics 2022; 14:pharmaceutics14122684. [PMID: 36559177 PMCID: PMC9783783 DOI: 10.3390/pharmaceutics14122684] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/07/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022] Open
Abstract
Tofacitinib, a Janus kinase 1 and 3 inhibitor, is used to treat rheumatoid arthritis. It is mainly metabolized by the cytochromes p450 (CYP) 3A1/2 and CYP2C11 in the liver. Chronic inflammation eventually leads to cirrhosis in patients with rheumatoid arthritis. Isosakuranetin (ISN), a component of Citrus aurantium L., has hepatoprotective effects in rats. This study was performed to determine the effects of ISN on the pharmacokinetics of tofacitinib in rats with N-dimethylnitrosamine-induced liver cirrhosis (LC). After intravenous administration of 10 mg/kg tofacitinib to control (CON), LC, and LC treated with ISN (LC-ISN) rats, the total area under the plasma concentration-time curves (AUC) from time zero to infinity increased by 158% in LC rats compared to those in CON rats; however, the AUC of LC-ISN rats decreased by 35.1% compared to that of LC rat. Similar patterns of AUC changes were observed in the LC and LC-ISN rats after oral administration of 20 mg/kg tofacitinib. These results can be attributed to decreased non-renal clearance (CLNR) and intestinal intrinsic clearance (CLint) in the LC rats and increased intestinal and hepatic CLint in the LC-ISN rats. Our findings imply that ISN treatment in LC rats restored the decrease in either CLNR or CLint, or both, through increased hepatic and intestinal expression of CYP3A1/2 and CYP2C11, which is regulated by the induction of pregnane X receptor (PXR) and constitutive androstane receptor (CAR).
Collapse
Affiliation(s)
- Sung Hun Bae
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, 206 Worldcup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
| | - Hyeon Gyeom Choi
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, 206 Worldcup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
| | - So Yeon Park
- Department of Biohealth Regulatory Science, Graduate School of Ajou University, 206 Worldcup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
| | - Sun-Young Chang
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, 206 Worldcup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
- Department of Biohealth Regulatory Science, Graduate School of Ajou University, 206 Worldcup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
| | - Hyoungsu Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, 206 Worldcup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
| | - So Hee Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, 206 Worldcup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
- Department of Biohealth Regulatory Science, Graduate School of Ajou University, 206 Worldcup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
- Correspondence: ; Tel.: +82-31-219-3451
| |
Collapse
|
9
|
Bae SH, Kim HS, Choi HG, Chang SY, Kim SH. Effects of Dextran Sulfate Sodium-Induced Ulcerative Colitis on the Disposition of Tofacitinib in Rats. Biomol Ther (Seoul) 2022; 30:510-519. [PMID: 35811297 PMCID: PMC9622310 DOI: 10.4062/biomolther.2022.049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 11/06/2022] Open
Abstract
Tofacitinib, a Janus kinase 1 and 3 inhibitor, is mainly metabolized by CYP3A1/2 and CYP2C11 in the liver. The drug has been approved for the chronic treatment of severe ulcerative colitis, a chronic inflammatory bowel disease. This study investigated the pharmacokinetics of tofacitinib in rats with dextran sulfate sodium (DSS)-induced ulcerative colitis. After 1-min of intravenous infusion of tofacitinib (10 mg/kg), the area under the plasma concentration-time curves from time zero to time infinity (AUC) of tofacitinib significantly increased by 92.3%. The time-averaged total body clearance decreased significantly by 47.7% in DSS rats compared with control rats. After the oral administration of tofacitinib (20 mg/kg), the AUC increased by 85.5% in DSS rats. These results could be due to decreased intrinsic clearance of the drug caused by the reduction of CYP3A1/2 and CYP2C11 in the liver and intestine of DSS rats. In conclusion, ulcerative colitis inhibited CYP3A1/2 and CYP2C11 in the liver and intestines of DSS rats and slowed the metabolism of tofacitinib, resulting in increased plasma concentrations of tofacitinib in DSS rats.
Collapse
Affiliation(s)
- Sung Hun Bae
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Hyo Sung Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Hyeon Gyeom Choi
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Sun-Young Chang
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - So Hee Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| |
Collapse
|
10
|
Laux J, Forster M, Riexinger L, Schwamborn A, Guezguez J, Pokoj C, Kudolo M, Berger LM, Knapp S, Schollmeyer D, Guse J, Burnet M, Laufer SA. Pharmacokinetic Optimization of Small Molecule Janus Kinase 3 Inhibitors to Target Immune Cells. ACS PHARMACOLOGY & TRANSLATIONAL SCIENCE 2022; 5:573-602. [DOI: 10.1021/acsptsci.2c00054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Julian Laux
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Michael Forster
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, DE Germany
| | - Laura Riexinger
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Anna Schwamborn
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Jamil Guezguez
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Christina Pokoj
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Mark Kudolo
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, DE Germany
| | - Lena M. Berger
- Structural Genomics Consortium, Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Stefan Knapp
- Structural Genomics Consortium, Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Dieter Schollmeyer
- Institute for Organic Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55099 Mainz, Germany
| | - Jan Guse
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Michael Burnet
- Synovo GmbH, Paul-Ehrlich-Straße 15, 72076 Tübingen, Germany
| | - Stefan A. Laufer
- Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, DE Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72076 Tübingen, Germany
- Tübingen Center for Academic Drug Discovery and Development (TüCAD2), 72076 Tübingen, Germany
| |
Collapse
|
11
|
Figueroa-Romero C, Monteagudo A, Murdock BJ, Famie JP, Webber-Davis IF, Piecuch CE, Teener SJ, Pacut C, Goutman SA, Feldman EL. Tofacitinib Suppresses Natural Killer Cells In Vitro and In Vivo: Implications for Amyotrophic Lateral Sclerosis. Front Immunol 2022; 13:773288. [PMID: 35197969 PMCID: PMC8859451 DOI: 10.3389/fimmu.2022.773288] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/18/2022] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal and incurable neurodegenerative disease with few therapeutic options. However, the immune system, including natural killer (NK) cells, is linked to ALS progression and may constitute a viable therapeutic ALS target. Tofacitinib is an FDA-approved immunomodulating small molecule which suppresses immune cell function by blocking proinflammatory cytokine signaling. This includes the cytokine IL-15 which is the primary cytokine associated with NK cell function and proliferation. However, the impact of tofacitinib on NK activation and cytotoxicity has not been thoroughly investigated, particularly in ALS. We therefore tested the ability of tofacitinib to suppress cytotoxicity and cytokine production in an NK cell line and in primary NK cells derived from control and ALS participants. We also investigated whether tofacitinib protected ALS neurons from NK cell cytotoxicity. Finally, we conducted a comprehensive pharmacokinetic study of tofacitinib in mice and tested the feasibility of administration formulated in chow. Success was assessed through the impact of tofacitinib on peripheral NK cell levels in mice. We found tofacitinib suppressed IL-15-induced activation as measured by STAT1 phosphorylation, cytotoxicity, pro-inflammatory gene expression, and pro-inflammatory cytokine secretion in both an NK cell line and primary NK cells. Furthermore, tofacitinib protected ALS neurons from NK cell-mediated cytotoxicity. In mice, we found tofacitinib bioavailability was 37% in both male and female mice; using these data we formulated mouse containing low and high doses of tofacitinib and found that the drug suppressed peripheral NK cell levels in a dose-dependent manner. These results demonstrate that tofacitinib can suppress NK cell function and may be a viable therapeutic strategy for ALS.
Collapse
Affiliation(s)
| | - Alina Monteagudo
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Benjamin J Murdock
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Joshua P Famie
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Ian F Webber-Davis
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Caroline E Piecuch
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Samuel J Teener
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Crystal Pacut
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Stephen A Goutman
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
12
|
Wiwatchaitawee K, Mekkawy AI, Quarterman JC, Naguib YW, Ebeid K, Geary SM, Salem AK. The MEK 1/2 inhibitor PD98059 exhibits synergistic anti-endometrial cancer activity with paclitaxel in vitro and enhanced tissue distribution in vivo when formulated into PAMAM-coated PLGA-PEG nanoparticles. Drug Deliv Transl Res 2021; 12:1684-1696. [PMID: 34635984 DOI: 10.1007/s13346-021-01065-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2021] [Indexed: 11/25/2022]
Abstract
Endometrial cancer is the most common gynecological cancer that affects the female reproductive organs. The standard therapy for EC for the past two decades has been chemotherapy and/or radiotherapy. PD98059 is a reversible MEK inhibitor that was found in these studies to increase the cytotoxicity of paclitaxel (PTX) against human endometrial cancer cells (Hec50co) in a synergistic and dose-dependent manner. Additionally, while PD98059 arrested Hec50co cells at the G0/G1 phase, and PTX increased accumulation of cells at the G2/M phase, the combination treatment increased accumulation at both the G0/G1 and G2/M phases at low PTX concentrations. We recently developed poly(lactide-co-glycolide) (PLGA) nanoparticles (NPs) modified with polyethylene glycol (PEG) and coated with polyamidoamine (PAMAM) (referred to here as PGM NPs) which have favorable biodistribution profiles in mice, compared to PD98059 solution. Here, in order to enhance tissue distribution of PD98059, PD98059-loaded PGM NPs were prepared and characterized. The average size, zeta potential, and % encapsulation efficiency (%EE) of these NPs was approximately 184 nm, + 18 mV, and 23%, respectively. The PD98059-loaded PGM NPs released ~ 25% of the total load within 3 days in vitro. In vivo murine studies revealed that the pharmacokinetics and biodistribution profile of intravenous (IV) injected PD98059 was improved when delivered as PD98059-loaded PGM NPs as opposed to soluble PD98059. Further investigation of the in vivo efficacy and safety of this formulation is expected to emphasize the potential of its clinical application in combination with commercial PTX formulations against different cancers.
Collapse
Affiliation(s)
- Kanawat Wiwatchaitawee
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, 52242, USA
| | - Aml I Mekkawy
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, 52242, USA
- Department of Pharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, Sohag University, Sohag, 82524, Egypt
| | - Juliana C Quarterman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, 52242, USA
| | - Youssef W Naguib
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, 52242, USA
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Manufacturing, Deraya University, New Minia City, 61768, Minia, Egypt
| | - Kareem Ebeid
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, 52242, USA
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Manufacturing, Deraya University, New Minia City, 61768, Minia, Egypt
| | - Sean M Geary
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, 52242, USA
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, 52242, USA.
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
13
|
Günaydın C, Önger ME, Avcı B, Bozkurt A, Terzi M, Bilge SS. Tofacitinib enhances remyelination and improves myelin integrity in cuprizone-induced mice. Immunopharmacol Immunotoxicol 2021; 43:790-798. [PMID: 34618622 DOI: 10.1080/08923973.2021.1986063] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
AIM Demyelination and subsequent remyelination are well-known mechanisms in multiple sclerosis (MS) pathology. Current research mainly focused on preventing demyelination or regulating the peripheral immune system to protect further damage to the central nervous system. However, information about another essential mechanism, remyelination, and its balance of the immune response within the central nervous system's boundaries is still limited. MATERIALS AND METHODS In this study, we tried to demonstrate the effect of the recently introduced Janus kinase (JAK)-signal transducer and activator of transcription (STAT) inhibitor, tofacitinib, on remyelination.Demyelination was induced by 6-week cuprizone administration, followed by 2-week tofacitinib (10, 30, and 100 mg/kg) treatment. RESULTS At the functional level, tofacitinib improved cuprizone-induced decline in motor coordination and muscle strength, which were assessed by rotarod and hanging wire tests. Tofacitinib also showed anti-inflammatory effect by alleviating the cuprizone-induced increase in the central levels of interferon-γ (IFN-γ), interleukin (IL)-6, IL-1β, and tumor necrosis alpha (TNF-α). Furthermore, tofacitinib also suppressed the cuprizone-induced increase in matrix metalloproteinases (MMP)-9 and MMP-2 levels. Additionally, cuprizone-induced loss of myelin integrity and myelin basic protein expression was inhibited by tofacitinib. At the molecular level, we also assessed phosphorylation of STAT-3 and STAT-5, and our data indicates tofacitinib suppressed cuprizone-induced phosphorylation in those proteins. CONCLUSION Our study highlights JAK/STAT inhibition provides beneficial effects on remyelination via inhibition of inflammatory cascade.
Collapse
Affiliation(s)
- Caner Günaydın
- Department of Pharmacology, School of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - M Emin Önger
- Department of Histology and Embryology, School of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Bahattin Avcı
- Department of Biochemistry, School of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Ayhan Bozkurt
- Department of Physiology, School of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Murat Terzi
- Department of Neurology, School of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - S Sırrı Bilge
- Department of Pharmacology, School of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| |
Collapse
|
14
|
Dose-independent pharmacokinetics of loganin in rats: effect of intestinal first-pass metabolism on bioavailability. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2021. [DOI: 10.1007/s40005-021-00546-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
15
|
Li Y, Sun Y, Wei S, Zhang L, Zong S. Development and evaluation of tofacitinib transdermal system for the treatment of rheumatoid arthritis in rats. Drug Dev Ind Pharm 2021; 47:878-886. [PMID: 33886401 DOI: 10.1080/03639045.2021.1916521] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
CONTEXT Tofacitinib tablet is approved for the treatment of rheumatoid arthritis (RA). However, tofacitinib (Tfc) faces extensive first-pass metabolism following oral administration. AIM To develop transdermal systems of Tfc and evaluate their efficacies against RA using Freund's Complete Adjuvant immunized arthritis rat model. METHODS These systems were prepared by solvent casting method and evaluated for texture, needle strength, skin penetrability, in vitro drug release, skin permeation, stability, and in vivo anti-arthritic activity. RESULTS AND DISCUSSION Transdermal patch (TS) showed smooth texture, good mechanical strength, slow-release, and slow permeation through the skin. Microneedle array (MNS) showed good needle strength, with required skin penetrability. MNS and TS showed 95% and 24% drug release, and 82% and 12% drug permeation, respectively in 4 h. The developed systems were found to be stable for 90 days at very stressful conditions, that is, 40 ± 2 °C and 75 ± 5% RH. MNS and TS both reduced arthritic scores (at p < 0.01 and p < 0.001 level, respectively) and the level of inflammatory cytokines (at p < 0.05 and p < 0.01 level, respectively) significantly as compared to that of the drug solution (DS). MNS and TS were found to be effective in restoring histological alterations (annum, synovial hyperplasia, synovial constriction, and cartilage and articular erosions) toward normal. CONCLUSION TS and MNS were found to be stable and effective for the treatment of arthritis and hence considered a good alternative for the treatment of RA with better clinical pertinence.
Collapse
Affiliation(s)
- Yanmei Li
- Department of Rheumatology and Immunology, Yantaishan Hospital, Yantai, China
| | - Yonghua Sun
- Department of Rheumatology and Immunology, Yantaishan Hospital, Yantai, China
| | - Shitong Wei
- Department of Rheumatology and Immunology, Yantaishan Hospital, Yantai, China
| | - Luyang Zhang
- Department of Rheumatology and Immunology, Yantaishan Hospital, Yantai, China
| | - Shihua Zong
- Department of Rheumatology and Immunology, Yantaishan Hospital, Yantai, China
| |
Collapse
|
16
|
Pharmacokinetic Drug Interaction between Tofacitinib and Voriconazole in Rats. Pharmaceutics 2021; 13:pharmaceutics13050740. [PMID: 34069798 PMCID: PMC8157262 DOI: 10.3390/pharmaceutics13050740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 12/14/2022] Open
Abstract
Fungal infections are prevalent in patients with immune diseases. Voriconazole, a triazole antifungal drug, inhibits the cytochromes CYP3A4 and CYP2C, and tofacitinib, a Janus kinase inhibitor for the treatment of rheumatoid arthritis, is metabolized by CYP3A4 and CYP2C19 in humans. Here, we investigated their interaction during simultaneous administration of both drugs to rats, either intravenously or orally. The area under the plasma concentration–time curve from time zero to time infinity (AUC) of tofacitinib was significantly greater, by 166% and 171%, respectively, and the time-averaged non-renal clearance (CLNR) of tofacitinib was significantly slower (59.5%) than that for tofacitinib alone. An in vitro metabolism study showed non-competitive inhibition of tofacitinib metabolism in the liver and intestine by voriconazole. The concentration/apparent inhibition constant (Ki) ratios of voriconazole were greater than two, indicating that the inhibition of tofacitinib metabolism could be due to the inhibition of the CYP3A1/2 and CYP2C11 enzymes by voriconazole. The pharmacokinetics of voriconazole were not affected by the co-administration of tofacitinib. In conclusion, the significantly greater AUC and slower CLNR of tofacitinib after intravenous and oral administration of both drugs were attributable to the non-competitive inhibition of tofacitinib metabolism via CYP3A1/2 and CYP2C11 by voriconazole in rats.
Collapse
|
17
|
Liu P, Wu S, Dong Z. Identification of the metabolites of tofacitinib in liver microsomes by liquid chromatography combined with high resolution mass spectrometry. Biomed Chromatogr 2021; 35:e5081. [PMID: 33522621 DOI: 10.1002/bmc.5081] [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: 12/09/2020] [Revised: 01/19/2021] [Accepted: 01/29/2021] [Indexed: 01/25/2023]
Abstract
Tofacitinib is an orally available Janus kinase inhibitor. The aim of this study was to investigate the metabolism of tofacitinib in mouse, rat, monkey, and human liver microsomes fortified with β-nicotinamide adenine dinucleotide phosphate tetrasodium salt and uridine diphosphate glucuronic acid. The biotransformation was executed at a temperature of 37°C for 60 min, and the samples were analyzed by ultra-high performance liquid chromatography combined with high-resolution mass spectrometry (UHPLC-HRMS) operated in positive electrospray ionization mode. The structures of the metabolites were elucidated according to their retention times, accurate masses, and MS/MS spectra. Under the current conditions, a total of 13 metabolites, including 1 glucuronide conjugate, were detected and structurally proposed. Oxygenation of the pyrrolopyrimidine ring, oxygenation of piperidine ring, N-demethylation, oxygenation of piperidine ring side chain, and glucuronidation were the primary metabolic pathways of tofacitinib. Among the tested species, tofacitinib showed significant species difference. Compared with other species, rat showed similar metabolic profiles to those of humans. The present study provides some new information regarding the metabolism of tofacitinib in animals and humans, which would bring us considerable benefits for the subsequent studies focusing on the pharmacological effect and toxicity of this drug.
Collapse
Affiliation(s)
- Ping Liu
- Department of Pharmacy, The Third Affiliated Hospital of Shandong First Medical University, The Fourth People's Hospital of Jinan, Jinan, China
| | - Shuang Wu
- Department of Pharmacy, The Third Affiliated Hospital of Shandong First Medical University, The Fourth People's Hospital of Jinan, Jinan, China
| | - Zhaoqin Dong
- Department of Pharmacy, The Third Affiliated Hospital of Shandong First Medical University, The Fourth People's Hospital of Jinan, Jinan, China
| |
Collapse
|
18
|
Huizing LRS, McDuffie J, Cuyckens F, van Heerden M, Koudriakova T, Heeren RMA, Vreeken RJ. Quantitative Mass Spectrometry Imaging to Study Drug Distribution in the Intestine Following Oral Dosing. Anal Chem 2021; 93:2144-2151. [DOI: 10.1021/acs.analchem.0c03956] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Lennart R. S. Huizing
- Maastricht MultiModal Molecular Imaging Institute, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - James McDuffie
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121, United States
| | - Filip Cuyckens
- Janssen Research & Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | | | - Tatiana Koudriakova
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121, United States
| | - Ron M. A. Heeren
- Maastricht MultiModal Molecular Imaging Institute, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Rob J. Vreeken
- Maastricht MultiModal Molecular Imaging Institute, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
- Janssen Research & Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| |
Collapse
|
19
|
Bae SH, Chang SY, Kim SH. Slower Elimination of Tofacitinib in Acute Renal Failure Rat Models: Contribution of Hepatic Metabolism and Renal Excretion. Pharmaceutics 2020; 12:pharmaceutics12080714. [PMID: 32751547 PMCID: PMC7464785 DOI: 10.3390/pharmaceutics12080714] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/20/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
Tofacitinib is a Jak inhibitor developed as a treatment for rheumatoid arthritis. Tofacitinib is metabolized mainly through hepatic CYP3A1/2, followed by CYP2C11. Rheumatoid arthritis tends to increase renal toxicity due to drugs used for long-term treatment. In this study, pharmacokinetic changes of tofacitinib were evaluated in rats with gentamicin (G-ARF) and cisplatin-induced acute renal failure (C-ARF). The time-averaged total body clearance (CL) of tofacitinib in G-ARF and C-ARF rats after 1-min intravenous infusion of 10 mg/kg was significantly decreased by 37.7 and 62.3%, respectively, compared to in control rats. This seems to be because the time-averaged renal clearance (CLR) was significantly lower by 69.5 and 98.6%, respectively, due to decreased creatinine clearance (CLCR). In addition, the time-averaged nonrenal clearance (CLNR) was also significantly lower by 33.2 and 57.4%, respectively, due to reduction in the hepatic CYP3A1/2 and CYP2C11 subfamily in G-ARF and C-ARF rats. After oral administration of tofacitinib (20 mg/kg) to G-ARF and C-ARF rats, both CLR and CLNR were also significantly decreased. In conclusion, an increase in area under plasma concentration-time curves from time zero to time infinity (AUC) of tofacitinib in G-ARF and C-ARF rats was due to the significantly slower elimination of tofacitinib contributed by slower hepatic metabolism and urinary excretion of the drug.
Collapse
Affiliation(s)
| | | | - So Hee Kim
- Correspondence: ; Tel.: +82-31-219-3451; Fax: +82-31-219-3435
| |
Collapse
|
20
|
Gwak EH, Yoo HY, Kim SH. Effects of Diabetes Mellitus on the Disposition of Tofacitinib, a Janus Kinase Inhibitor, in Rats. Biomol Ther (Seoul) 2020; 28:361-369. [PMID: 32209733 PMCID: PMC7327145 DOI: 10.4062/biomolther.2020.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/14/2020] [Accepted: 02/21/2020] [Indexed: 12/21/2022] Open
Abstract
Tofacitinib, a Janus kinase inhibitor, was developed for the treatment of rheumatoid arthritis. Recently, it has been associated with an increased change in arthritis development in patients with diabetes. Herein, we evaluated the pharmacokinetics of tofacitinib after intravenous (10 mg/kg) and oral (20 mg/kg) administration to rats with streptozotocin-induced diabetes mellitus and control rats. Following intravenous administration of tofacitinib to rats with streptozotocin-induced diabetes mellitus, area under the plasma concentration-time curve from time zero to infinity of tofacitinib was significantly smaller (33.6%) than that of control rats. This might be due to the faster hepatic intrinsic clearance (112%) caused by an increase in the hepatic cytochrome P450 (CYP) 3A1(23) and the faster hepatic blood flow rate in rats with streptozotocin-induced diabetes mellitus than in control rats. Following oral administration, area under the plasma concentration-time curve from time zero to infinity of tofacitinib was also significantly smaller (55.5%) in rats with streptozotocin-induced diabetes mellitus than that in control rats. This might be due to decreased absorption caused by the higher expression of P-glycoprotein and the faster intestinal metabolism caused by the higher expression of intestinal CYP3A1(23), which resulted in the decreased bioavailability of tofacitinib (33.0%) in rats with streptozotocin-induced diabetes mellitus. In summary, our findings indicate that diabetes mellitus affects the absorption and metabolism of tofacitinib, causing faster metabolism and decreased intestinal absorption in rats with streptozotocin-induced diabetes mellitus.
Collapse
Affiliation(s)
- Eun Hye Gwak
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Hee Young Yoo
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - So Hee Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| |
Collapse
|
21
|
Simple determination and quantification of tofacitinib, a JAK inhibitor, in rat plasma, urine and tissue homogenates by HPLC and its application to a pharmacokinetic study. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2020. [DOI: 10.1007/s40005-020-00490-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
22
|
Veeravalli V, Dash RP, Thomas JA, Babu RJ, Madgula LMV, Srinivas NR. Critical Assessment of Pharmacokinetic Drug–Drug Interaction Potential of Tofacitinib, Baricitinib and Upadacitinib, the Three Approved Janus Kinase Inhibitors for Rheumatoid Arthritis Treatment. Drug Saf 2020; 43:711-725. [DOI: 10.1007/s40264-020-00938-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|