1
|
Jeon JH, Jeon SY, Baek YJ, Park CE, Choi MK, Han YT, Song IS. Pharmacokinetics and Enterohepatic Circulation of 2-(Quinoline-8-carboxamido)benzoic Acid (2-QBA) in Mice. Pharmaceutics 2024; 16:934. [PMID: 39065631 PMCID: PMC11279551 DOI: 10.3390/pharmaceutics16070934] [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: 06/15/2024] [Revised: 07/06/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
The quinoline alkaloid 2-(quinoline-8-carboxamido)benzoic acid (2-QBA), which is isolated from Aspergillus sp. SCSIO06786, a deep sea-derived fungus, has been suggested as a therapeutic candidate for the treatment of Parkinson's disease. We developed an analytical method for 2-QBA using a liquid chromatography-tandem mass spectrometry (LC-MS/MS) in mouse plasma, in which a protein precipitation method for the sample preparation of 2-QBA in mouse plasma was used due to its simplicity and good extraction recovery rates (80.49-97.56%). The linearity of the calibration standard sample, inter- and intraday precision and accuracy, and stability of three quality control samples were suitable based on the assessment criteria and the lower limit of quantification (LLOQ) of the 2-QBA was 1 ng/mL. A pharmacokinetic study of 2-QBA was performed in mice divided into oral (2.0, 5.0, and 15 mg/kg) and intravenous (0.5 and 1.0 mg/kg) administration groups. The absolute oral bioavailability (BA) range of 2-QBA was calculated as 68.3-83.7%. Secondary peaks were observed at approximately 4-8 h after the oral administration of 2-QBA at all doses. The elimination half-life of the orally administered 2-QBA was significantly longer than that of the intravenous 2-QBA. In addition, glucuronide metabolites of 2-QBA were identified. They were transformed into 2-QBA using the β-glucuronidase treatment. Furthermore, the 2-QBA was readily absorbed from the jejunum to lower ileum. Taken together, the secondary peaks could be explained by the enterohepatic circulation of 2-QBA. In conclusion, the reabsorption of orally administered 2-QBA could contribute to the high oral BA of 2-QBA and could be beneficial for the efficacy of 2-QBA. Moreover, the simple and validated analytical method for 2-QBA using LC-MS/MS was applied to the pharmacokinetic study and BA assessments of 2-QBA in mice and would be helpful for subsequent pharmacokinetic studies, as well as for evaluations of the toxicokinetics and pharmacokinetic-pharmacodynamic correlation of 2-QBA to assess its potential as a drug.
Collapse
Affiliation(s)
- Ji-Hyeon Jeon
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, Vessel-Organ Interaction Research Center (VOICE), College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - So-Yeon Jeon
- College of Pharmacy, Dankook University, Cheon-an 31116, Republic of Korea; (S.-Y.J.); (Y.-J.B.); (C.-E.P.); (M.-K.C.)
| | - Yeon-Ju Baek
- College of Pharmacy, Dankook University, Cheon-an 31116, Republic of Korea; (S.-Y.J.); (Y.-J.B.); (C.-E.P.); (M.-K.C.)
| | - Chan-E Park
- College of Pharmacy, Dankook University, Cheon-an 31116, Republic of Korea; (S.-Y.J.); (Y.-J.B.); (C.-E.P.); (M.-K.C.)
| | - Min-Koo Choi
- College of Pharmacy, Dankook University, Cheon-an 31116, Republic of Korea; (S.-Y.J.); (Y.-J.B.); (C.-E.P.); (M.-K.C.)
| | - Young Taek Han
- College of Pharmacy, Dankook University, Cheon-an 31116, Republic of Korea; (S.-Y.J.); (Y.-J.B.); (C.-E.P.); (M.-K.C.)
| | - Im-Sook Song
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, Vessel-Organ Interaction Research Center (VOICE), College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea;
| |
Collapse
|
2
|
Ma Y, Zhao Y, Luo M, Jiang Q, Liu S, Jia Q, Bai Z, Wu F, Xie J. Advancements and challenges in pharmacokinetic and pharmacodynamic research on the traditional Chinese medicine saponins: a comprehensive review. Front Pharmacol 2024; 15:1393409. [PMID: 38774213 PMCID: PMC11106373 DOI: 10.3389/fphar.2024.1393409] [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: 02/29/2024] [Accepted: 04/12/2024] [Indexed: 05/24/2024] Open
Abstract
Recent research on traditional Chinese medicine (TCM) saponin pharmacokinetics has revealed transformative breakthroughs and challenges. The multicomponent nature of TCM makes it difficult to select representative indicators for pharmacokinetic studies. The clinical application of saponins is limited by their low bioavailability and short half-life, resulting in fluctuating plasma concentrations. Future directions should focus on novel saponin compounds utilizing colon-specific delivery and osmotic pump systems to enhance oral bioavailability. Optimizing drug combinations, such as ginsenosides with aspirin, shows therapeutic potential. Rigorous clinical validation is essential for practical applications. This review emphasizes a transformative era in saponin research, highlighting the need for clinical validation. TCM saponin pharmacokinetics, guided by traditional principles, are in development, utilizing multidisciplinary approaches for a comprehensive understanding. This research provides a theoretical basis for new clinical drugs and supports rational clinical medication.
Collapse
Affiliation(s)
- Yuhan Ma
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Yongxia Zhao
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Mingxia Luo
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Qin Jiang
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Sha Liu
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Qi Jia
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Zhixun Bai
- Organ Transplant Center, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Faming Wu
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Jian Xie
- School of Pharmacy, Zunyi Medical University, Zunyi, China
- School of Preclinical Medicine, Zunyi Medical University, Zunyi, China
| |
Collapse
|
3
|
Xie L, Zhao YX, Zheng Y, Li XF. The pharmacology and mechanisms of platycodin D, an active triterpenoid saponin from Platycodon grandiflorus. Front Pharmacol 2023; 14:1148853. [PMID: 37089949 PMCID: PMC10117678 DOI: 10.3389/fphar.2023.1148853] [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: 01/20/2023] [Accepted: 03/28/2023] [Indexed: 04/09/2023] Open
Abstract
Chinese doctors widely prescribed Platycodon grandiflorus A. DC. (PG) to treat lung carbuncles in ancient China. Modern clinical experiences have demonstrated that PG plays a crucial role in treating chronic pharyngitis, plum pneumonia, pneumoconiosis, acute and chronic laryngitis, and so forth. Additionally, PG is a food with a long history in China, Japan, and Korea. Furthermore, Platycodin D (PLD), an oleanane-type triterpenoid saponin, is one of the active substances in PG. PLD has been revealed to have anti-inflammatory, anti-viral, anti-oxidation, anti-obesity, anticoagulant, spermicidal, anti-tumor etc., activities. And the mechanism of the effects draws lots of attention, with various signaling pathways involved in these processes. Additionally, research on PLD's pharmacokinetics and extraction processes is under study. The bioavailability of PLD could be improved by being prescribed with Glycyrrhiza uralensis Fisch. or by creating a new dosage form. PLD has been recently considered to have the potential to be a solubilizer or an immunologic adjuvant. Meanwhile, PLD was discovered to have hemolytic activity correlated. PLD has broad application prospects and reveals practical pharmacological activities in pre-clinical research. The authors believe that these activities of PLD contribute to the efficacy of PG. What is apparent is that the clinical translation of PLD still has a long way to go. With the help of modern technology, the scope of clinical applications of PLD is probable to be expanded from traditional applications to new fields.
Collapse
Affiliation(s)
| | | | | | - Xiao-Fang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
4
|
Enhanced bioavailability and hepatoprotective effect of silymarin by preparing silymarin-loaded solid dispersion formulation using freeze-drying method. Arch Pharm Res 2022; 45:743-760. [PMID: 36178580 DOI: 10.1007/s12272-022-01407-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 09/13/2022] [Indexed: 11/02/2022]
Abstract
This study aimed to develop a solid dispersion formulation of silymarin (Silymarin-SD) using freeze-drying method to enhance its oral bioavailability (BA) by inhibiting the intestinal first-pass effect and increasing its solubility and permeability. Silymarin-SD formulation (i.e., silymarin:tween 80:hydroxypropyl cellulose (HPC) = 1:1:3 (w/w/w) significantly increased silymarin permeability in the duodenum, jejunum, and ileum by decreasing the efflux ratio of silymarin and by inhibiting silymarin-glucuronidation activity, in which tween 80 played a crucial role. As a result, orally administered Silymarin-SD formulation increased plasma silymarin concentrations and decreased silymarin-glucuronide in rats compared with silymarin alone and silymmarin:D-α-tocopherol polyethylene glycol 1000 succinate (1:1, w/w) formulation. In addition to modulating intestinal first-pass effect, Silymarin-SD formulation showed a significantly higher cumulative dissolution for 120 min compared with that of silymarin from the physical mixture (PM) of the same composition as Silymarin-SD and silymarin alone; the relative BA of silymarin-SD increased to 215% and 589% compared with silymarin-PM and silymarin alone, respectively. This could be attributed to the amorphous status of the Silymarin-SD formulation without chemical interaction with excipients, such as tween 80 and HPC. Moreover, the hepatoprotective effect of Silymarin-SD in acetaminophen-induced acute hepatotoxicity, as estimated from the alanine aminotransferase and aspartate aminotransferase values, was superior to that of silymarin. In conclusion, the increase in the dissolution rate and intestinal permeability of silymarin, and the inhibition of silymarin-glucuronidation by the Silymarin-SD formulation, prepared using tween 80 and HPC, increased its plasma concentration and resulted in a superior hepatoprotective effect compared to silymarin.
Collapse
|
5
|
Chen H, Li G, Liu Y, Lang Y, Yang W, Zhang W, Liang X. Jiegeng Decoction Potentiates the Anticancer Efficacy of Paclitaxel in vivo and in vitro. Front Pharmacol 2022; 13:827520. [PMID: 35281908 PMCID: PMC8914467 DOI: 10.3389/fphar.2022.827520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/31/2022] [Indexed: 11/30/2022] Open
Abstract
Paclitaxel (PTX) has been the first-line treatment for lung cancer; however, its clinical use is limited due to multidrug resistance (MDR) and adverse effects. Thus, there is an urgent need to explore agents that can enhance the anticancer efficacy of PTX by reducing drug resistance and adverse reactions. Jiegeng decoction (JG) was used as the meridian guide drug and adjuvant drug in treatment of lung cancer. However, the mechanism of adjuvant effect was unclear. The aim of this study was to determine whether JG could potentiate the anticancer effect of PTX. Tissue distribution of PTX was detected using HPLC-MS/MS. The anti-lung cancer effect of the combination of PTX and JG in Lewis lung cancer C57BL/6J mice was evaluated based on the body weight and tumor-inhibition rate. PTX concentration in tumors was determined using HPLC-MS and in vivo imaging. Biochemical indices were detected using biochemical analyzer and ELISA. The anticancer mechanism of the PTX-JG combination in A549/PTX cells was elucidated based on cell proliferation, annexin V-FITC apoptosis assay, and western blotting. Tissue distribution analysis showed that the distribution of PTX increased in the lungs, liver, and heart upon administering the combination of PTX and JG. JG remarkably enhanced the anticancer effect of PTX by increasing the red blood cell and platelet counts; increasing hemoglobin, interleukin (IL)-2, and tumor necrosis factor-α levels; increasing CD4+T cells and the CD4+/CD8+ ratio; and decreasing IL-10 levels. JG administration led to the increased distribution of PTX at the tumor lesion sites and also potentiated the anticancer effect of PTX by inhibiting tumor cell proliferation and promoting apoptosis. Moreover, JG reversed PTX resistance by inhibiting the expression of lung resistance-related proteins, multiresistance protein 1, P-glycoprotein, and breast cancer-resistant protein. Furthermore, the combination of JG and PTX decreased alanine aminotransferase and aspartate aminotransferase levels and did not affect creatine kinase-MB levels. Therefore, our discovery suggests that JG increased the anticancer effect of PTX by downregulating the MDR-related protein and demonstrated a synergistic enhancement of immunity. Thus, the combination of PTX with JG shows potential in the management of lung cancer owing to its synergistic and detoxifying effects.
Collapse
Affiliation(s)
- Haifang Chen
- Jiangxi University of Chinese Medicine, Nanchang, China
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Guofeng Li
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Ye Liu
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yifan Lang
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Wuliang Yang
- Jiangxi University of Chinese Medicine, Nanchang, China
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Wugang Zhang
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Xinli Liang
- Jiangxi University of Chinese Medicine, Nanchang, China
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
6
|
Li Q, Yang T, Zhao S, Zheng Q, Li Y, Zhang Z, Sun X, Liu Y, Zhang Y, Xie J. Distribution, biotransformation, pharmacological effects, metabolic mechanism and safety evaluation of Platycodin D:A comprehensive review. Curr Drug Metab 2022; 23:21-29. [PMID: 35114917 DOI: 10.2174/1389200223666220202090137] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/05/2021] [Accepted: 12/31/2021] [Indexed: 11/22/2022]
Abstract
Platycodonis Radix (Jiegeng), the dried root of Platycodon grandiflorum, is a traditional herb used as both medicine and food. Its clinical application for the treatment of cough, phlegm, sore throat, pulmonary and respiratory diseases has been thousands of years in China. Platycodin D is the main active ingredient in Platycodonis Radix, which belongs to the family of pentacyclic triterpenoid saponins because it contains an oleanolane type aglycone linked with double sugar chains. Modern pharmacology has demonstrated that Platycodin D displays various biological activities, such as analgesics, expectoration and cough suppression, promoting weight loss, anti-tumor and immune regulation, suggesting that Platycodin D has the potential to be a drug candidate and an interesting target as a natural product for clinical research. In this review, the distribution and biotransformation, pharmacological effects, metabolic mechanism and safety evaluation of Platycodin D are summarized to lay the foundation for further studies.
Collapse
Affiliation(s)
- Qianqian Li
- College of Traditional Chinese Pharmacy, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China
| | - Tan Yang
- College of Traditional Chinese Pharmacy, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China
| | - Shuang Zhao
- College of Traditional Chinese Pharmacy, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China
| | - Qifeng Zheng
- College of Traditional Chinese Pharmacy, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China
| | - Yaxin Li
- Department of Chemistry, Center for Gene Regulation in Health and Diseases, Cleveland State University, Cleveland, OH, 44115, USA
| | - Zhiyuan Zhang
- College of Traditional Chinese Pharmacy, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China
| | - Xiuyan Sun
- College of Traditional Chinese Pharmacy, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China
| | - Yan Liu
- Department of Pharmacy, Weifang People\'s Hospital, Weifang, 261041, People's Republic of China
| | - Yanqing Zhang
- College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, People's Republic of China
| | - Junbo Xie
- College of Traditional Chinese Pharmacy, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China
| |
Collapse
|
7
|
Jeon JH, Lee J, Park JH, Lee CH, Choi MK, Song IS. Effect of Lactic Acid Bacteria on the Pharmacokinetics and Metabolism of Ginsenosides in Mice. Pharmaceutics 2021; 13:1496. [PMID: 34575573 PMCID: PMC8469489 DOI: 10.3390/pharmaceutics13091496] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 12/23/2022] Open
Abstract
This study aims to investigate the effect of lactic acid bacteria (LAB) on in vitro and in vivo metabolism and the pharmacokinetics of ginsenosides in mice. When the in vitro fermentation test of RGE with LAB was carried out, protopanaxadiol (PPD) and protopanaxadiol (PPD), which are final metabolites of ginsenosides but not contained in RGE, were greatly increased. Compound K (CK), ginsenoside Rh1 (GRh1), and GRg3 also increased by about 30%. Other ginsenosides with a sugar number of more than 2 showed a gradual decrease by fermentation with LAB for 7 days, suggesting the involvement of LAB in the deglycosylation of ginsenosides. Incubation of single ginsenoside with LAB produced GRg3, CK, and PPD with the highest formation rate and GRd, GRh2, and GF with the lower rate among PPD-type ginsenosides. Among PPT-type ginsenosides, GRh1 and PPT had the highest formation rate. The amoxicillin pretreatment (20 mg/kg/day, twice a day for 3 days) resulted in a significant decrease in the fecal recovery of CK, PPD, and PPT through the blockade of deglycosylation of ginsenosides after single oral administrations of RGE (2 g/kg) in mice. The plasma concentrations of CK, PPD, and PPT were not detectable without change in GRb1, GRb2, and GRc in this group. LAB supplementation (1 billion CFU/2 g/kg/day for 1 week) after the amoxicillin treatment in mice restored the ginsenoside metabolism and the plasma concentrations of ginsenosides to the control level. In conclusion, the alterations in the gut microbiota environment could change the ginsenoside metabolism and plasma concentrations of ginsenosides. Therefore, the supplementation of LAB with oral administrations of RGE would help increase plasma concentrations of deglycosylated ginsenosides such as CK, PPD, and PPT.
Collapse
Affiliation(s)
- Ji-Hyeon Jeon
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, Vessel-Organ Interaction Research Center (VOICE), Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea; (J.-H.J.); (J.L.); (J.-H.P.)
| | - Jaehyeok Lee
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, Vessel-Organ Interaction Research Center (VOICE), Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea; (J.-H.J.); (J.L.); (J.-H.P.)
| | - Jin-Hyang Park
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, Vessel-Organ Interaction Research Center (VOICE), Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea; (J.-H.J.); (J.L.); (J.-H.P.)
| | - Chul-Haeng Lee
- College of Pharmacy, Dankook University, Cheon-an 31116, Korea;
| | - Min-Koo Choi
- College of Pharmacy, Dankook University, Cheon-an 31116, Korea;
| | - Im-Sook Song
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, Vessel-Organ Interaction Research Center (VOICE), Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea; (J.-H.J.); (J.L.); (J.-H.P.)
| |
Collapse
|
8
|
Shen J, Zhu Y, Zhou B, Kong L, Jin Y, Zhang D, Cao Z, Ji J, Li J. In vitro and in vivo evaluation of a water-in-oil microemulsion of platycodin D. Arch Pharm (Weinheim) 2021; 354:e2000497. [PMID: 33844326 DOI: 10.1002/ardp.202000497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 11/11/2022]
Abstract
Platycodin D (PD) is the active metabolite of Platycodon grandiflorum. The main purpose of this study was to develop and evaluate a water-in-oil (W/O) microemulsion formulation of PD (PD-ME). The PD-ME was successfully prepared by the water titration method at K m = 2, to draw the pseudoternary phase diagrams. Physical characterization including the particle size, pH, refractive index, average viscosity, and polydispersity index (PDI) was performed. The in vivo characteristics were evaluated by intestinal permeability and pharmacokinetic studies. The optimized microemulsion formulation consisted of 100 mg/ml PD aqueous solution, soybean phospholipids, ethanol, and oleic acid (27:39:19:15, w/w). The average viscosity, pH, droplet size, PDI, and zeta potential of the PD-ME were 78.65 ± 0.13 cPa•s, 5.70 ± 0.05, 30.46 ± 0.20 nm, 0.33 ± 0.00, and -3.13 mV, respectively. The drug concentration of the PD-ME was 26.3 ± 0.6 mg/ml. The PD-ME showed significantly higher apparent permeability coefficients than PD (p < .01). The pharmacokinetic studies showed that the PD-ME had significantly higher values of T 1/2 (2.26-fold), AUC0-24h (area under the curve; 1.65-fold), and MRT0-24h (1.58-fold) than PD (p < .01). It can be seen that W/O ME presents a strategy with great promise for enhancing the intestinal permeability and better oral absorption of drugs with high polarity and poor permeability.
Collapse
Affiliation(s)
- Jinyang Shen
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang, Jiangsu, China
- Department of Pharmacy, Jiangsu Ocean University, Lianyungang, Jiangsu, China
| | - Yuexia Zhu
- Department of Pharmacy, Jiangsu Ocean University, Lianyungang, Jiangsu, China
| | - Bingxue Zhou
- Department of Pharmacy, Jiangsu Ocean University, Lianyungang, Jiangsu, China
| | - Li Kong
- Department of Pharmacy, Jiangsu Ocean University, Lianyungang, Jiangsu, China
| | - Ye Jin
- Department of Pharmacy, Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou, Jiangsu, China
| | - Di Zhang
- Department of Pharmacy, Jiangsu Ocean University, Lianyungang, Jiangsu, China
| | - Zhiling Cao
- Department of Pharmacy, Jiangsu Ocean University, Lianyungang, Jiangsu, China
| | - Jing Ji
- Department of Pharmacy, Jiangsu Ocean University, Lianyungang, Jiangsu, China
| | - Jiaojiao Li
- Department of Pharmacy, Jiangsu Ocean University, Lianyungang, Jiangsu, China
| |
Collapse
|
9
|
Song IS, Nam SJ, Jeon JH, Park SJ, Choi MK. Enhanced Bioavailability and Efficacy of Silymarin Solid Dispersion in Rats with Acetaminophen-Induced Hepatotoxicity. Pharmaceutics 2021; 13:pharmaceutics13050628. [PMID: 33925040 PMCID: PMC8146637 DOI: 10.3390/pharmaceutics13050628] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
We evaluated the bioavailability, liver distribution, and efficacy of silymarin-D-α-tocopherol polyethylene glycol 1000 succinate (TPGS) solid dispersion (silymarin-SD) in rats with acetaminophen-induced hepatotoxicity (APAP) compared with silymarin alone. The solubility of silybin, the major and active component of silymarin, in the silymarin-SD group increased 23-fold compared with the silymarin group. The absorptive permeability of silybin increased by 4.6-fold and its efflux ratio decreased from 5.5 to 0.6 in the presence of TPGS. The results suggested that TPGS functioned as a solubilizing agent and permeation enhancer by inhibiting efflux pump. Thus, silybin concentrations in plasma and liver were increased in the silymarin-SD group and liver distribution increased 3.4-fold after repeated oral administration of silymarin-SD (20 mg/kg as silybin) for five consecutive days compared with that of silymarin alone (20 mg/kg as silybin). Based on higher liver silybin concentrations in the silymarin-SD group, the therapeutic effects of silymarin-SD in hepatotoxic rats were evaluated and compared with silymarin administration only. Elevated alanine aminotransferase, aspartate aminotransferase, and alkaline phosphatase levels were significantly decreased by silymarin-SD, silymarin, and TPGS treatments, but these decreases were much higher in silymarin-SD animals than in those treated with silymarin or TPGS. In conclusion, silymarin-SD (20 mg/kg as silybin, three times per day for 5 days) exhibited hepatoprotective properties toward hepatotoxic rats and these properties were superior to silymarin alone, which may be attributed to increased solubility, enhanced intestinal permeability, and increased liver distribution of the silymarin-SD formulation.
Collapse
Affiliation(s)
- Im-Sook Song
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, Vessel-Organ Interaction Research Center (VOICE), Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea; (S.-J.N.); (J.-H.J.)
- Correspondence: (I.-S.S.); (M.-K.C.); Tel.: +82-53-950-8575 (I.-S.S.); +82-41-550-1438 (M.-K.C.); Fax: +82-53-950-8557 (I.-S.S.)
| | - So-Jeong Nam
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, Vessel-Organ Interaction Research Center (VOICE), Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea; (S.-J.N.); (J.-H.J.)
| | - Ji-Hyeon Jeon
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, Vessel-Organ Interaction Research Center (VOICE), Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Korea; (S.-J.N.); (J.-H.J.)
| | - Soo-Jin Park
- College of Korean Medicine, Daegu Haany University, Daegu 38610, Korea;
| | - Min-Koo Choi
- College of Pharmacy, Dankook University, Cheon-an 31116, Korea
- Correspondence: (I.-S.S.); (M.-K.C.); Tel.: +82-53-950-8575 (I.-S.S.); +82-41-550-1438 (M.-K.C.); Fax: +82-53-950-8557 (I.-S.S.)
| |
Collapse
|
10
|
Shin YC, Shin W, Koh D, Wu A, Ambrosini YM, Min S, Eckhardt SG, Fleming RYD, Kim S, Park S, Koh H, Yoo TK, Kim HJ. Three-Dimensional Regeneration of Patient-Derived Intestinal Organoid Epithelium in a Physiodynamic Mucosal Interface-on-a-Chip. MICROMACHINES 2020; 11:E663. [PMID: 32645991 PMCID: PMC7408321 DOI: 10.3390/mi11070663] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/03/2020] [Accepted: 07/05/2020] [Indexed: 12/29/2022]
Abstract
The regeneration of the mucosal interface of the human intestine is critical in the host-gut microbiome crosstalk associated with gastrointestinal diseases. The biopsy-derived intestinal organoids provide genetic information of patients with physiological cytodifferentiation. However, the enclosed lumen and static culture condition substantially limit the utility of patient-derived organoids for microbiome-associated disease modeling. Here, we report a patient-specific three-dimensional (3D) physiodynamic mucosal interface-on-a-chip (PMI Chip) that provides a microphysiological intestinal milieu under defined biomechanics. The real-time imaging and computational simulation of the PMI Chip verified the recapitulation of non-linear luminal and microvascular flow that simulates the hydrodynamics in a living human gut. The multiaxial deformations in a convoluted microchannel not only induced dynamic cell strains but also enhanced particle mixing in the lumen microchannel. Under this physiodynamic condition, an organoid-derived epithelium obtained from the patients diagnosed with Crohn's disease, ulcerative colitis, or colorectal cancer independently formed 3D epithelial layers with disease-specific differentiations. Moreover, co-culture with the human fecal microbiome in an anoxic-oxic interface resulted in the formation of stochastic microcolonies without a loss of epithelial barrier function. We envision that the patient-specific PMI Chip that conveys genetic, epigenetic, and environmental factors of individual patients will potentially demonstrate the pathophysiological dynamics and complex host-microbiome crosstalk to target a patient-specific disease modeling.
Collapse
Affiliation(s)
- Yong Cheol Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
| | - Woojung Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
| | - Domin Koh
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
| | - Alexander Wu
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
| | - Yoko M. Ambrosini
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
| | - Soyoun Min
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
| | - S. Gail Eckhardt
- Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA; (S.G.E.); (R.Y.D.F.)
| | - R. Y. Declan Fleming
- Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA; (S.G.E.); (R.Y.D.F.)
- Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Seung Kim
- Severance Fecal Microbiota Transplantation Center, Severance Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Korea; (S.K.); (S.P.); (H.K.)
| | - Sowon Park
- Severance Fecal Microbiota Transplantation Center, Severance Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Korea; (S.K.); (S.P.); (H.K.)
| | - Hong Koh
- Severance Fecal Microbiota Transplantation Center, Severance Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Korea; (S.K.); (S.P.); (H.K.)
| | - Tae Kyung Yoo
- Department of Computer Art, College of Art and Technology, Chung-Ang University, Seoul 06974, Korea;
| | - Hyun Jung Kim
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
- Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA; (S.G.E.); (R.Y.D.F.)
- Department of Medical Engineering, College of Medicine, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
11
|
Bailly C, Vergoten G. Proposed mechanisms for the extracellular release of PD-L1 by the anticancer saponin platycodin D. Int Immunopharmacol 2020; 85:106675. [PMID: 32531711 DOI: 10.1016/j.intimp.2020.106675] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/02/2020] [Accepted: 06/02/2020] [Indexed: 12/11/2022]
Abstract
Platycodin D (PTD) is an oleanane-type terpenoid saponin, isolated from the plant Platycodon grandiflorus. PTD displays multiple pharmacological effects, notably significant anticancer activities in vitro and in vivo. Recently, PTD was shown to trigger the extracellular release of the immunologic checkpoint glycoprotein PD-L1. The reduction of PD-L1 expression at the surface of cancer cells leads to interleukin-2 secretion and T cells activation. In the present review, we have analyzed the potential origin of this atypical PTD-induced PD-L1 release to propose a mechanistic explanation. For that, we considered all published scientific information, as well as the physicochemical characteristics of the natural product (a modeling analysis of PTD and the related saponin β -escin is provided). On this basis, we raise the hypothesis that the capacity of PTD to induce PD-L1 extracellular release derives from two main mechanisms: (i) a drug-promoted shedding of membrane PD-L1 by metalloproteases or more likely, (ii) a cholesterol binding-related effect, that would lead to perturbation of membrane raft domains, limiting the recruitment of proteins like TLR4. The drug-induced membrane effects (frequently observed with saponin drugs), associated with a production of interferon-γ,can favor the release of proteins like PD-L1 into membrane vesicles. Our analysis supports the hypothesis that PTD is a cholesterol-dependent lipid raft-modulating agent able to promote the formation of PD-L1 containing extracellular vesicles. The anticancer potential of PTD and its capacity to modulate the functioning of the PD-1/PD-L1 checkpoint should be further considered.
Collapse
Affiliation(s)
| | - Gérard Vergoten
- University of Lille, Inserm, U995 - LIRIC - Lille Inflammation Research International Center, ICPAL, 3 rue du Professeur Laguesse, BP-83, F-59006 Lille, France
| |
Collapse
|
12
|
Enhanced Intestinal Permeability and Plasma Concentration of Metformin in Rats by the Repeated Administration of Red Ginseng Extract. Pharmaceutics 2019; 11:pharmaceutics11040189. [PMID: 31003498 PMCID: PMC6523382 DOI: 10.3390/pharmaceutics11040189] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 04/16/2019] [Indexed: 01/10/2023] Open
Abstract
We aimed to assess the potential herb-drug interactions between Korean red ginseng extract (RGE) and metformin in rats in terms of the modulation of metformin transporters, such as organic cation transporter (Oct), multiple toxin and extrusion protein (Mate), and plasma membrane monoamine transporter (Pmat). Single treatment of RGE did not inhibit the in vitro transport activity of OCT1/2 up to 500 µg/mL and inhibited MATE1/2-K with high IC50 value (more than 147.8 µg/mL), suggesting that concomitant used of RGE did not directly inhibit OCT- and MATE-mediated metformin uptake. However, 1-week repeated administration of RGE (1.5 g/kg/day) (1WRA) to rats showed different alterations in mRNA levels of Oct1 depending on the tissue type. RGE increased intestinal Oct1 but decreased hepatic Oct1. However, neither renal Oct1/Oct2 nor Mate1/Pmat expression in duodenum, jejunum, ileum, liver, and kidney were changed in 1WRA rats. RGE repeated dose also increased the intestinal permeability of metformin; however, the permeability of 3-O-methyl-d-glucose and Lucifer yellow was not changed in 1WRA rats, suggesting that the increased permeability of metformin by multiple doses of RGE is substrate-specific. On pharmacokinetic analysis, plasma metformin concentrations following intravenous injection were not changed in 1WRA, consistent with no significant change in renal Oct1, Oct2, and mate1. Repeated doses of RGE for 1 week significantly increased the plasma concentration of metformin, with increased half-life and urinary excretion of metformin following oral administration of metformin (50 mg/kg), which could be attributed to the increased absorption of metformin. In conclusion, repeated administration of RGE showed in vivo pharmacokinetic herb-drug interaction with metformin, with regard to its plasma exposure and increased absorption in rats. These results were consistent with increased intestinal Oct1 and its functional consequence, therefore, the combined therapeutic efficacy needs further evaluation before the combination and repeated administration of RGE and metformin, an Oct1 substrate drug.
Collapse
|
13
|
Choi YA, Song IS, Choi MK. Pharmacokinetic Drug-Drug Interaction and Responsible Mechanism between Memantine and Cimetidine. Pharmaceutics 2018; 10:pharmaceutics10030119. [PMID: 30082658 PMCID: PMC6161283 DOI: 10.3390/pharmaceutics10030119] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/31/2018] [Accepted: 08/03/2018] [Indexed: 11/27/2022] Open
Abstract
A sensitive and simple chromatography-tandem mass spectrometry (LC-MS/MS) method was developed to evaluate memantine in rat plasma. Memantine and propranolol (internal standard) in rat plasma was extracted using a methanol precipitation method. The standard curve value was 0.2–1000 ng/mL and selectivity, linearity, inter-day and intra-day accuracy and precision were within acceptance criteria. Using this validated method, drug-drug interactions between memantine and cimetidine was measured following co-administration of memantine and cimetidine intravenously and orally. Plasma exposure of memantine was increased by 1.6- and 3.0-fold by co-medication with cimetidine intravenously and orally, respectively. It suggested that the drug interaction occurred during the gut absorption process, which was consistent with the results showing that the intestinal permeability of memantine in the presence of cimetidine was 3.2-fold greater than that of memantine alone. Inhibition of cimetidine on hepatic elimination of memantine rather than renal excretion was also attributed to the drug-drug interaction between memantine and cimetidine, which explained the decreased clearance of memantine by co-medication with cimetidine. In conclusion, the newly developed simple and sensitive LC-MS/MS analytical method was applied to investigate the pharmacokinetic drug-drug interactions of memantine. Plasma exposure of memantine by co-administration with cimetidine was increased because of its enhanced intestinal permeability and the decreased metabolic activity of memantine.
Collapse
Affiliation(s)
- Young A Choi
- College of Pharmacy, Dankook University, Cheon-an 31116, Korea.
| | - Im-Sook Song
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Min-Koo Choi
- College of Pharmacy, Dankook University, Cheon-an 31116, Korea.
| |
Collapse
|