1
|
Michiba K, Watanabe K, Imaoka T, Nakai D. Recent Advances in the Gastrointestinal Complex in Vitro Model for ADME Studies. Pharmaceutics 2023; 16:37. [PMID: 38258048 PMCID: PMC10819272 DOI: 10.3390/pharmaceutics16010037] [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: 10/30/2023] [Revised: 12/06/2023] [Accepted: 12/16/2023] [Indexed: 01/24/2024] Open
Abstract
Intestinal absorption is a complex process involving the permeability of the epithelial barrier, efflux transporter activity, and intestinal metabolism. Identifying the key factors that govern intestinal absorption for each investigational drug is crucial. To assess and predict intestinal absorption in humans, it is necessary to leverage appropriate in vitro systems. Traditionally, Caco-2 monolayer systems and intestinal Ussing chamber studies have been considered the 'gold standard' for studying intestinal absorption. However, these methods have limitations that hinder their universal use in drug discovery and development. Recently, there has been an increasing number of reports on complex in vitro models (CIVMs) using human intestinal organoids derived from intestinal tissue specimens or iPSC-derived enterocytes plated on 2D or 3D in microphysiological systems. These CIVMs provide a more physiologically relevant representation of key ADME-related proteins compared to conventional in vitro methods. They hold great promise for use in drug discovery and development due to their ability to replicate the expressions and functions of these proteins. This review highlights recent advances in gut CIVMs employing intestinal organoid model systems compared to conventional methods. It is important to note that each CIVM should be tailored to the investigational drug properties and research questions at hand.
Collapse
Affiliation(s)
- Kazuyoshi Michiba
- Drug Metabolism & Pharmacokinetics Research Laboratory, Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan; (K.W.); (T.I.); (D.N.)
| | | | | | | |
Collapse
|
2
|
Li L, Qi Z, Han S, Li X, Liu B, Liu Y. Advances and Applications of Metal-Organic Framework Nanomaterials as Oral Delivery Carriers: A Review. Mini Rev Med Chem 2022; 22:2564-2580. [PMID: 35362373 DOI: 10.2174/1389557522666220330152145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/27/2022] [Accepted: 02/15/2022] [Indexed: 11/22/2022]
Abstract
Oral administration is a commonly used, safe, and patient-compliant method of drug delivery. However, due to the multiple absorption barriers in the gastrointestinal tract (GIT), the oral bioavailability of many drugs is low, resulting in a limited range of applications for oral drug delivery. Nanodrug delivery systems have unique advantages in overcoming the multiple barriers to oral absorption and improving the oral bioavailability of encapsulated drugs. Metal-organic frameworks (MOFs) are composed of metal ions and organic linkers assembled by coordination chemistry. Unlike other nanomaterials, nanoscale metal-organic frameworks (nano-MOFs, NMOFs) are increasingly popular for drug delivery systems (DDSs) due to their tunable pore size and easily modified surfaces. This paper summarizes the literature on MOFs in pharmaceutics included in SCI for the past ten years. Then, the GIT structure and oral drug delivery systems are reviewed, and the advantages, challenges, and solution strategies possessed by oral drug delivery systems are discussed. Importantly, two major classes of MOFs suitable for oral drug delivery systems are summarized, and various representative MOFs as oral drug carriers are evaluated in the context of oral drug delivery systems. Finally, the challenges faced by DDSs in the development of MOFs, such as biostability, biosafety, and toxicity, are examined.
Collapse
Affiliation(s)
- Li Li
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, 110000, China;
- Institute of Forensic Expertise, Liaoning University, Shenyang, 110000, China
| | - Zhaorui Qi
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, 110000, China
| | - Shasha Han
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, 110000, China
| | - Xurui Li
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, 110000, China
| | - Bingmi Liu
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, 110000, China;
- Institute of Forensic Expertise, Liaoning University, Shenyang, 110000, China
| | - Yu Liu
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, 110000, China;
- Institute of Forensic Expertise, Liaoning University, Shenyang, 110000, China
| |
Collapse
|
3
|
Nielsen RB, Holm R, Pijpers I, Snoeys J, Nielsen UG, Nielsen CU. Oral etoposide and zosuquidar bioavailability in rats: Effect of co-administration and in vitro-in vivo correlation of P-glycoprotein inhibition. Int J Pharm X 2021; 3:100089. [PMID: 34977557 PMCID: PMC8683663 DOI: 10.1016/j.ijpx.2021.100089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 07/03/2021] [Indexed: 11/01/2022] Open
Abstract
P-glycoprotein inhibitors, like zosuquidar, have widely been used to study the role of P-glycoprotein in oral absorption. Still, systematic studies on the inhibitor dose-response relationship on intestinal drug permeation are lacking. In the present study, we investigated the effect of 0.79 nM-2.5 μM zosuquidar on etoposide permeability across Caco-2 cell monolayers. We also investigated etoposide pharmacokinetics after oral or IV administration to Sprague Dawley rats with co-administration of 0.063–63 mg/kg zosuquidar, as well as the pharmacokinetics of zosuquidar itself. Oral zosuquidar bioavailability was 2.6–4.2%, while oral etoposide bioavailability was 5.5 ± 0.9%, which increased with increasing zosuquidar doses to 35 ± 5%. The intestinal zosuquidar concentration required to induce a half-maximal increase in bioavailability was estimated to 180 μM. In contrast, the IC50 of zosuquidar on etoposide permeability in vitro was only 5–10 nM, and a substantial in vitro-in vivo discrepancy of at least four orders of magnitude was thereby identified. Overall, the present study provides valuable insights for future formulation development that applies fixed dose combinations of P-glycoprotein inhibitors to increase the absorption of poorly permeable P-glycoprotein substrate drugs.
Collapse
|
4
|
Drug Disposition in the Lower Gastrointestinal Tract: Targeting and Monitoring. Pharmaceutics 2021; 13:pharmaceutics13020161. [PMID: 33530468 PMCID: PMC7912393 DOI: 10.3390/pharmaceutics13020161] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/20/2022] Open
Abstract
The increasing prevalence of colonic diseases calls for a better understanding of the various colonic drug absorption barriers of colon-targeted formulations, and for reliable in vitro tools that accurately predict local drug disposition. In vivo relevant incubation conditions have been shown to better capture the composition of the limited colonic fluid and have resulted in relevant degradation and dissolution kinetics of drugs and formulations. Furthermore, drug hurdles such as efflux transporters and metabolising enzymes, and the presence of mucus and microbiome are slowly integrated into drug stability- and permeation assays. Traditionally, the well characterized Caco-2 cell line and the Ussing chamber technique are used to assess the absorption characteristics of small drug molecules. Recently, various stem cell-derived intestinal systems have emerged, closely mimicking epithelial physiology. Models that can assess microbiome-mediated drug metabolism or enable coculturing of gut microbiome with epithelial cells are also increasingly explored. Here we provide a comprehensive overview of the colonic physiology in relation to drug absorption, and review colon-targeting formulation strategies and in vitro tools to characterize colonic drug disposition.
Collapse
|
5
|
Michiba K, Maeda K, Kurimori K, Enomoto T, Shimomura O, Takeuchi T, Nishiyama H, Oda T, Kusuhara H. Characterization of the Human Intestinal Drug Transport with Ussing Chamber System Incorporating Freshly Isolated Human Jejunum. Drug Metab Dispos 2021; 49:84-93. [PMID: 33087448 DOI: 10.1124/dmd.120.000138] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 10/02/2020] [Indexed: 02/13/2025] Open
Abstract
Intestinal permeability is a critical factor for orally administered drugs. It can be facilitated by uptake transporters or limited by efflux transporters and metabolic enzymes in the intestine. The present study aimed to characterize the Ussing chamber system incorporating human intestinal tissue as an in vitro model for investigating the impact of intestinal uptake/efflux transporters on the intestinal absorption of substrate drugs in humans. We confirmed the functions of major intestinal uptake/efflux drug transporters in freshly isolated human jejunum sections by demonstrating a significant decrease in the mucosal uptake of cefadroxil (peptide transporter 1) and methotrexate (proton-coupled folate transporter), mucosal-to-serosal permeability of ribavirin (concentrative nucleoside transporters/equilibrative nucleoside transporters), and serosal-to-mucosal permeability of P-glycoprotein and breast cancer resistance protein substrates in the presence of their typical inhibitors. The mucosal-to-serosal apparent permeability coefficients (Papp) of 19 drugs, including substrates of drug transporters and cytochrome P450 3A, ranged from 0.60 × 10-6 to 29 × 10-6 cm/s and showed a good correlation with reported fraction of an oral dose that enters the gut wall and passes into the portal circulation with escaping intestinal metabolism (FaFg) values in humans. Furthermore, the Papp values for cefadroxil, methotrexate, and ribavirin in the presence of the corresponding transporter inhibitors underestimated the FaFg of these drugs, which clearly showed that intestinal uptake transporters facilitate their intestinal absorption in humans. In conclusion, the functions of major intestinal uptake/efflux drug transporters could be maintained in freshly isolated human jejunum sections. The Ussing chamber system incorporating human intestinal tissue would be useful for evaluating the impact of intestinal uptake/efflux transporters on the intestinal absorption of various types of drugs in humans. SIGNIFICANCE STATEMENT: Although previous studies have predicted the intestinal absorption of drugs in humans using the Ussing chamber system incorporating human intestinal tissue, there is little systematic information about drug transport mediated by multiple transporters in this system. We confirmed the functions of major intestinal uptake/efflux transporters in freshly isolated human jejunum sections and demonstrated that the mucosal-to-serosal apparent permeability coefficient of various types of drugs showed a good correlation with reported human FaFg values.
Collapse
Affiliation(s)
- Kazuyoshi Michiba
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (K.K., T.E., O.S., T.O.); and Tsukuba Human Tissue Biobank Center, University of Tsukuba Hospital, Ibaraki, Japan (T.T., H.N.)
| | - Kazuya Maeda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (K.K., T.E., O.S., T.O.); and Tsukuba Human Tissue Biobank Center, University of Tsukuba Hospital, Ibaraki, Japan (T.T., H.N.)
| | - Ko Kurimori
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (K.K., T.E., O.S., T.O.); and Tsukuba Human Tissue Biobank Center, University of Tsukuba Hospital, Ibaraki, Japan (T.T., H.N.)
| | - Tsuyoshi Enomoto
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (K.K., T.E., O.S., T.O.); and Tsukuba Human Tissue Biobank Center, University of Tsukuba Hospital, Ibaraki, Japan (T.T., H.N.)
| | - Osamu Shimomura
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (K.K., T.E., O.S., T.O.); and Tsukuba Human Tissue Biobank Center, University of Tsukuba Hospital, Ibaraki, Japan (T.T., H.N.)
| | - Tomoyo Takeuchi
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (K.K., T.E., O.S., T.O.); and Tsukuba Human Tissue Biobank Center, University of Tsukuba Hospital, Ibaraki, Japan (T.T., H.N.)
| | - Hiroyuki Nishiyama
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (K.K., T.E., O.S., T.O.); and Tsukuba Human Tissue Biobank Center, University of Tsukuba Hospital, Ibaraki, Japan (T.T., H.N.)
| | - Tatsuya Oda
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (K.K., T.E., O.S., T.O.); and Tsukuba Human Tissue Biobank Center, University of Tsukuba Hospital, Ibaraki, Japan (T.T., H.N.)
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (K.Mi., K.Ma., H.K.); Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan (K.K., T.E., O.S., T.O.); and Tsukuba Human Tissue Biobank Center, University of Tsukuba Hospital, Ibaraki, Japan (T.T., H.N.)
| |
Collapse
|
6
|
Formulation of a Phenol-Rich Extract from Unripe Olives ( Olea europaea L.) in Microemulsion to Improve Its Solubility and Intestinal Permeability. Molecules 2020; 25:molecules25143198. [PMID: 32668791 PMCID: PMC7397150 DOI: 10.3390/molecules25143198] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/03/2020] [Accepted: 07/09/2020] [Indexed: 12/14/2022] Open
Abstract
The beneficial properties of phenolic compounds from Olea europaea L. are well-known. An olive extract (OE) was prepared from unripe olives (Moraiolo cultivar). The study aimed to formulate OE into a microemulsion (ME) in oral dosage form. OE was extracted from olives with EtOH:H2O (80:20) and characterized by HPLC-DAD. ME composition was stated by a solubility and pseudo-ternary diagram. The ME was chemically and physically characterized, and its stability at 4 °C was analyzed for three months. The ability of the formulation to ameliorate the solubility and the intestinal permeability of OE was evaluated by a Parallel Artificial Membrane Permeability Assay (PAMPA) assay and Caco-2 cells. The total phenolic content of the extract was 39% w/w. The main constituent was oleuropein (31.0%), together with ligstroside (3.1%) and verbascoside (2.4%). The ME was prepared using Capryol 90 as the oily phase, and Cremophor EL and Transcutol (2:1) as surfactant and co-surfactant, respectively. ME droplet size was 14.03 ± 1.36 nm, PdI 0.20 ± 0.08, ζ-potential −1.16 ± 0.48. Stability of ME was confirmed for at least three months. The formulation was loaded with 35 mg/mL of OE, increasing the solubility of the extract by about four times. The enhanced permeability of OE was evaluated by PAMPA, as demonstrated by the Pe value (1.44 ± 0.83 × 10−6 cm/s for OE hydroalcoholic solution, 3.74 ± 0.34 × 10−6 cm/s for OE-ME). Caco-2 cell transport studies confirmed the same results: Papp was 16.14 ± 0.05 × 10−6 cm/s for OE solution and 26.99 ± 0.45 × 10−6 cm/s for OE-ME. ME proved to be a suitable formulation for oral delivery.
Collapse
|
7
|
Al-Ali AAA, Sandra L, Versweyveld D, Pijpers I, Dillen L, Vermeulen A, Snoeys J, Holm R, Nielsen CU. High-dose etoposide formulations do not saturate intestinal P-glycoprotein: Development, stability, and pharmacokinetics in Sprague-Dawley rats. Int J Pharm 2020; 583:119399. [DOI: 10.1016/j.ijpharm.2020.119399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/28/2020] [Accepted: 04/30/2020] [Indexed: 01/05/2023]
|
8
|
Zheng C, Li Y, Peng Z, He X, Tao J, Ge L, Sun Y, Wu Y. A composite nanocarrier to inhibit precipitation of the weakly basic drug in the gastrointestinal tract. Drug Deliv 2020; 27:712-722. [PMID: 32397763 PMCID: PMC7269033 DOI: 10.1080/10717544.2020.1760402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
For weakly basic drugs, the sharp decrease of drug solubility and the following drug precipitation after drugs transferring from the gastric fluid to the intestinal fluid in the gastrointestinal (GI) tract is a main reason for the poor oral bioavailability of drugs. Here, an anticoagulant dabigatran etexilate (DE) was used as a model drug, and a composite nanocarrier system of DE was developed to improve the drug dissolution by decreasing the drug leakage in the stomach and inhibiting the drug precipitation in the intestinal tract. With the encapsulation of drugs in nanocarriers, the precipitation percentage of DE in composite nanocarriers was 22.25 ± 3.88% in simulated intestinal fluid, which was far below that of the commercial formulation. Moreover, the relative bioavailability of DE-loaded composite nanocarriers (456.58%) was greatly enhanced and the peak of its activated partial thromboplastin time was also significantly prolonged (p < .01) compared with the commercial formulation, indicating that the anticoagulant effect of DE was effectively improved. Therefore, the designed composite nanocarrier system of DE presents great potentials in improving the therapeutic efficiency and expanding the clinical applications of poorly water-soluble weakly basic drugs.
Collapse
Affiliation(s)
- Chunli Zheng
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Yun Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Zhen Peng
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Xinyi He
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Juan Tao
- School of Pharmacy and Traditional Chinese Pharmacy, Jiangsu College of Nursing, Huaian, China
| | - Liang Ge
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Yixin Sun
- Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yunkai Wu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
9
|
Talegaonkar S, Bhattacharyya A. Potential of Lipid Nanoparticles (SLNs and NLCs) in Enhancing Oral Bioavailability of Drugs with Poor Intestinal Permeability. AAPS PharmSciTech 2019; 20:121. [PMID: 30805893 DOI: 10.1208/s12249-019-1337-8] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 02/04/2019] [Indexed: 11/30/2022] Open
Abstract
Lipid-based drug delivery systems has become a popular choice for oral delivery of lipophilic drugs with dissolution rate limited oral absorption. Lipids are known to enhance oral bioavailability of poorly water-soluble drugs in multiple ways like facilitating dissolution as micellar solution, enhancing the lymphatic uptake and acting as inhibitors of efflux transporters. Lipid nanoparticles are matrix type lipid-based carrier systems which can effectively encapsulate both lipophilic and hydrophilic drugs. Lipid nanoparticles namely solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) are versatile drug delivery system and can be used for multiple routes of delivery like parenteral, topical, ocular, transdermal, and oral. Lipid nanoparticles are particularly attractive vehicles for peroral delivery of drugs with oral bioavailability problems as they are composed of lipid excipients which are cheap, easily available, and non-toxic; manufacturing technique is simple and readily scalable for large-scale production; the formulations provide controlled release of active components and have no stability issue. A large number of drugs have been incorporated into lipid nanoparticles with the objective of overcoming their poor oral bioavailability. This review tries to assess the potential of lipid nanoparticles for enhancing the oral bioavailability of drugs with permeability limited oral absorption such as drugs belonging to class IV of Biopharmaceutic Classification System (BCS) and protein and peptide drugs and also discusses the mechanism behind the bioavailability enhancement and safety issues related to such delivery systems.
Collapse
|
10
|
Enright EF, Govindarajan K, Darrer R, MacSharry J, Joyce SA, Gahan CGM. Gut Microbiota-Mediated Bile Acid Transformations Alter the Cellular Response to Multidrug Resistant Transporter Substrates in Vitro: Focus on P-glycoprotein. Mol Pharm 2018; 15:5711-5727. [PMID: 30388019 DOI: 10.1021/acs.molpharmaceut.8b00875] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pharmacokinetic research at the host-microbe interface has been primarily directed toward effects on drug metabolism, with fewer investigations considering the absorption process. We previously demonstrated that the transcriptional expression of genes encoding intestinal transporters involved in lipid translocation are altered in germ-free and conventionalized mice possessing distinct bile acid signatures. It was consequently hypothesized that microbial bile acid metabolism, which is the deconjugation and dehydroxylation of the bile acid steroid nucleus by gut bacteria, may impact upon drug transporter expression and/or activity and potentially alter drug disposition. Using a panel of three human intestinal cell lines (Caco-2, T84, and HT-29) that differ in basal transporter expression level, bile acid conjugation-, and hydroxylation-status was shown to influence the transcription of genes encoding several major influx and efflux transporter proteins. We further investigated if these effects on transporter mRNA would translate to altered drug disposition and activity. The results demonstrated that the conjugation and hydroxylation status of the bile acid steroid nucleus can influence the cellular response to multidrug resistance (MDR) substrates, a finding that did not directly correlate with directionality of gene or protein expression. In particular, we noted that the cytotoxicity of cyclosporine A was significantly augmented in the presence of the unconjugated bile acids deoxycholic acid (DCA) and chenodeoxycholic acid (CDCA) in P-gp positive cell lines, as compared to their taurine/glycine-conjugated counterparts, implicating P-gp in the molecular response. Overall this work identifies a novel mechanism by which gut microbial metabolites may influence drug accumulation and suggests a potential role for the microbial bile acid-deconjugating enzyme bile salt hydrolase (BSH) in ameliorating multidrug resistance through the generation of bile acid species with the capacity to access and inhibit P-gp ATPase. The physicochemical property of nonionization is suggested to underpin the preferential ability of unconjugated bile acids to attenuate the efflux of P-gp substrates and to sensitize tumorigenic cells to cytotoxic therapeutics in vitro. This work provides new impetus to investigate whether perturbation of the gut microbiota, and thereby the bile acid component of the intestinal metabolome, could alter drug pharmacokinetics in vivo. These findings may additionally contribute to the development of less toxic P-gp modulators, which could overcome MDR.
Collapse
Affiliation(s)
- Elaine F Enright
- School of Pharmacy , ‡APC Microbiome Ireland , §School of Biochemistry and Cell Biology , ∥School of Microbiology , ⊥School of Medicine , University College Cork , Cork , Ireland
| | | | | | | | | | - Cormac G M Gahan
- School of Pharmacy , ‡APC Microbiome Ireland , §School of Biochemistry and Cell Biology , ∥School of Microbiology , ⊥School of Medicine , University College Cork , Cork , Ireland
| |
Collapse
|
11
|
P-glycoprotein expression in the gastrointestinal tract of male and female rats is influenced differently by food. Eur J Pharm Sci 2018; 123:569-575. [DOI: 10.1016/j.ejps.2018.08.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 12/17/2022]
|
12
|
Schumacher-Klinger A, Fanous J, Merzbach S, Weinmüller M, Reichart F, Räder AFB, Gitlin-Domagalska A, Gilon C, Kessler H, Hoffman A. Enhancing Oral Bioavailability of Cyclic RGD Hexa-peptides by the Lipophilic Prodrug Charge Masking Approach: Redirection of Peptide Intestinal Permeability from a Paracellular to Transcellular Pathway. Mol Pharm 2018; 15:3468-3477. [PMID: 29976060 DOI: 10.1021/acs.molpharmaceut.8b00466] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Hydrophilic peptides constitute most of the active peptides. They mostly permeate via tight junctions (paracellular pathway) in the intestine. This permeability mechanism restricts the magnitude of their oral absorption and bioavailability. We hypothesized that concealing the hydrophilic residues of the peptide using the lipophilic prodrug charge masking approach (LPCM) can improve the bioavailability of hydrophilic peptides. To test this hypothesis, a cyclic N-methylated hexapeptide containing Arg-Gly-Asp (RGD) and its prodrug derivatives, masking the Arg and Asp charged side chains, were synthesized. The library was evaluated for intestinal permeability in vitro using the Caco-2 model. Further investigation of metabolic stability ex vivo models in rat plasma, brush border membrane vesicles (BBMVs), and isolated CYP3A4 microsomes and pharmacokinetic studies was performed on a selected peptide and its prodrug (peptide 12). The parent drug analogues were found to have a low permeability rate in vitro, corresponding to atenolol, a marker for paracellular permeability. Moreover, palmitoyl carnitine increased the Papp of peptide 12 by 4-fold, indicating paracellular permeability. The Papp of the prodrug derivatives was much higher than that of their parent peptides. For instance, the Papp of the prodrug 12P was 20-fold higher than the Papp of peptide 12 in the apical to basolateral (AB) direction. Whereas the permeability in the opposite direction (BA of the Caco-2 model) was significantly faster than the Papp AB, indicating the involvement of an efflux system. These results were corroborated when verapamil, a P-gp inhibitor, was added to the Caco-2 model and increased the Papp AB of prodrug 12P by 3-fold. The prodrug 12P was stable in the BBMVs environment, yet degraded quickly (less than 5 min) in the plasma into the parent peptide 12. Pharmacokinetic studies in rats showed an increase in the bioavailability of peptide 12 > 70-fold (from 0.58 ± 0.11% to 43.8 ± 14.9%) after applying the LPCM method to peptide 12 and converting it to the prodrug 12P. To conclude, the LPCM approach converted the absorption mechanism of the polar peptides from a paracellular to transcellular pathway that tremendously affects their oral bioavailability. The LPCM method provides a solution for the poor bioavailability of RGD cyclohexapeptides and paves the way for other active hydrophilic and charged peptides with poor oral bioavailability.
Collapse
Affiliation(s)
- Adi Schumacher-Klinger
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine , The Hebrew University of Jerusalem , P.O. Box 12065, Jerusalem 91120 , Israel
| | - Joseph Fanous
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine , The Hebrew University of Jerusalem , P.O. Box 12065, Jerusalem 91120 , Israel
| | - Shira Merzbach
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine , The Hebrew University of Jerusalem , P.O. Box 12065, Jerusalem 91120 , Israel
| | - Michael Weinmüller
- Institute for Advanced Study and Center of Integrated Protein Science, Department Chemie , Technische Universität München , Lichtenbergstrasse 4 , 85748 Garching , Germany
| | - Florian Reichart
- Institute for Advanced Study and Center of Integrated Protein Science, Department Chemie , Technische Universität München , Lichtenbergstrasse 4 , 85748 Garching , Germany
| | - Andreas F B Räder
- Institute for Advanced Study and Center of Integrated Protein Science, Department Chemie , Technische Universität München , Lichtenbergstrasse 4 , 85748 Garching , Germany
| | - Agata Gitlin-Domagalska
- Institute of Chemistry , The Hebrew University of Jerusalem, Edmond Safra Campus, Givat Ram Campus, The Hebrew University , Jerusalem 91904 , Israel
| | - Chaim Gilon
- Institute of Chemistry , The Hebrew University of Jerusalem, Edmond Safra Campus, Givat Ram Campus, The Hebrew University , Jerusalem 91904 , Israel
| | - Horst Kessler
- Institute for Advanced Study and Center of Integrated Protein Science, Department Chemie , Technische Universität München , Lichtenbergstrasse 4 , 85748 Garching , Germany
| | - Amnon Hoffman
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine , The Hebrew University of Jerusalem , P.O. Box 12065, Jerusalem 91120 , Israel
| |
Collapse
|
13
|
Taccola C, Cartot-Cotton S, Valente D, Barneoud P, Aubert C, Boutet V, Gallen F, Lochus M, Nicolic S, Dodacki A, Smirnova M, Cisternino S, Declèves X, Bourasset F. High brain distribution of a new central nervous system drug candidate despite its P-glycoprotein-mediated efflux at the mouse blood-brain barrier. Eur J Pharm Sci 2018; 117:68-79. [DOI: 10.1016/j.ejps.2018.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/21/2017] [Accepted: 02/05/2018] [Indexed: 11/28/2022]
|
14
|
Xu J, Lin Y, Boulas P, Peterson ML. Low colonic absorption drugs: risks and opportunities in the development of oral extended release products. Expert Opin Drug Deliv 2017; 15:197-211. [PMID: 28988504 DOI: 10.1080/17425247.2018.1389889] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Currently numerous drugs have been observed with lower colonic absorption than small intestine absorption, which can significantly impact in vivo performance of their oral extended release (ER) products. AREAS COVERED We reviewed over 300 publications, patents, book chapters, and commercial reports of drug products from regulatory agencies for low colonic absorption (LCA) drugs and critical findings are discussed. The focuses of this article are (1) current findings on the causes of low colonic absorption to support early assessment of LCA candidates, and (2) current knowledge on successful ER strategies and technical platforms used for LCA drugs in commercial drug products to facilitate oral ER product development. EXPERT OPINION Colonic drug absorption is one of the critical considerations in successful development of oral ER products. The root causes of low colonic absorption in many LCA drugs are still unclear. It is recommended to evaluate colonic drug absorption of drug candidate at early stage of oral ER product development. After evaluation, the selection of a formulation platform to develop an oral ER product needs to be carefully considered for LCA drugs. Based on the current commercial oral ER formulation platforms for LCA drugs, compounds are first divided into five types (I-V) and different ER formulation approaches with higher success rate are recommended for each type.
Collapse
Affiliation(s)
- Jin Xu
- a Pharmaceutical Development , Biogen Inc , Cambridge , MA , USA
| | - Yiqing Lin
- a Pharmaceutical Development , Biogen Inc , Cambridge , MA , USA
| | - Pierre Boulas
- a Pharmaceutical Development , Biogen Inc , Cambridge , MA , USA
| | | |
Collapse
|
15
|
Tremblay S, Alloway RR. Clinical Evaluation of Modified Release and Immediate Release Tacrolimus Formulations. AAPS JOURNAL 2017; 19:1332-1347. [PMID: 28717926 DOI: 10.1208/s12248-017-0119-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 06/27/2017] [Indexed: 12/21/2022]
Abstract
The science of drug delivery has evolved considerably and has led to the development of multiple sustained release formulations. Each of these formulations can present particular challenges in terms of clinical evaluation and necessitate careful study to identify their optimal use in practice. Tacrolimus is an immunosuppressive agent that is widely used in organ transplant recipients. However, it is poorly soluble, has an unpredictable pharmacokinetic profile subject to important genetic polymorphisms and drug-drug interactions, and has a narrow therapeutic index. For these reasons, it represents an agent that could benefit from modified release formulations to overcome these limitations. The objective of this review is to discuss the clinical evaluation of immediate and modified release tacrolimus formulations in renal transplant recipients. Clinical trials from early development of immediate release tacrolimus to formulation-specific post-marketing trials of modified release tacrolimus formulations are reviewed with an emphasis on key elements relating to trial design end endpoint assessment. Particular elements that can be addressed with formulation alterations, such as pharmacokinetics, pharmacogenomics, and toxicity and corresponding clinical evaluations are discussed. In addition, current knowledge gaps in the clinical evaluation of immediate and modified release tacrolimus formulations are discussed to highlight potential avenues for the future development of different tacrolimus formulations with outcomes relevant to the regulators, the transplant community, and to transplant recipients. This review shows that new formulations may alter tacrolimus bioavailability, alleviate certain adverse events while potentially enhancing patient convenience.
Collapse
Affiliation(s)
- Simon Tremblay
- Department of Surgery, Division of Transplantation, University of Cincinnati College of Medicine, 231 Albert Sabin Way, ML0519, Cincinnati, OH, 45267, USA. .,Department of Environmental Health, Division of Epidemiology, University of Cincinnati College of Medicine, 231 Albert Sabin Way, ML0519, Cincinnati, OH, 45267, USA.
| | - Rita R Alloway
- Department of Internal Medicine, Division of Nephrology, Kidney C.A.R.E Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
16
|
Hermann KF, Neuhaus CS, Micallef V, Wagner B, Hatibovic M, Aschmann HE, Paech F, Alvarez-Sanchez R, Krämer SD, Belli S. Kinetics of lipid bilayer permeation of a series of ionisable drugs and their correlation with human transporter-independent intestinal permeability. Eur J Pharm Sci 2017; 104:150-161. [PMID: 28366650 DOI: 10.1016/j.ejps.2017.03.040] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/21/2017] [Accepted: 03/29/2017] [Indexed: 12/28/2022]
Abstract
For low molecular weight drugs, lipid bilayer permeation is considered the major route for in vivo cell barrier passage. We recently introduced a fluorescence assay with liposomes to determine permeation kinetics of ionisable compounds across the lipid bilayer by monitoring drug-induced pH changes inside the liposomes. Here, we determined the permeability coefficients (PFLipP, FLipP for "Fluorescence Liposomal Permeability") across 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) bilayers of 35 ionisable drugs at pH6.0 and compared them to available in vivo human jejunal permeability (Peff) data. PFLipP values were furthermore compared with published Caco-2 cell permeability coefficients (PCaco-2), permeability coefficients determined with the parallel artificial membrane permeability assay (PAMPA) and with log D (pH6.0). The log PFLipP, corrected for predicted para-cellular diffusion, and log PCaco-2 correlated best with log Peff, with similar adjusted R2 (0.75 and 0.74, n=12). Our results suggest that transporter-independent intestinal drug absorption is predictable from liposomal permeability.
Collapse
Affiliation(s)
- Katharina F Hermann
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
| | - Claudia S Neuhaus
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
| | - Virgine Micallef
- Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Björn Wagner
- Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Maja Hatibovic
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
| | - Hélène E Aschmann
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
| | - Franziska Paech
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland
| | - Rubén Alvarez-Sanchez
- Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Stefanie D Krämer
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 4, CH-8093 Zurich, Switzerland.
| | - Sara Belli
- Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland.
| |
Collapse
|
17
|
Pérez-Pitarch A, Guglieri-López B, Nacher A, Merino V, Merino-Sanjuan M. Levofloxacin effect on erlotinib absorption. Evaluation of the interaction in undernutrition situations through population pharmacokinetic analysis in rats. Biopharm Drug Dispos 2017; 38:315-325. [PMID: 28099756 DOI: 10.1002/bdd.2065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 11/16/2016] [Accepted: 01/16/2017] [Indexed: 11/09/2022]
Abstract
The main objective of this study was to develop a pharmacokinetic model in order to describe the intestinal absorption of erlotinib in rat and to quantify the interaction of levofloxacin on this process in well- and under-nourished rats. Absorption studies were performed in male Wistar rats. Concentration-time profiles in proximal and distal intestine were analysed through non-linear mixed effect modelling using the NONMEM software version 7.3. Simulations were performed in order to explore the influence of covariates on the apparent absorption rate constant. A passive absorption and an active secretion process best-described erlotinib absorption from lumen to enterocyte. The developed model indicates that levofloxacin exerts an inhibition on erlotinib efflux transporters of the gut epithelium. Undernourishment proved to significantly decrease the maximum capacity of the secretion process. Simulations evidenced that erlotinib absorption only takes place at high enough drug concentrations to overcome the effect of efflux transporters. On the other hand, when levofloxacin is present in the intestinal lumen of undernourished rats, erlotinib drug absorption takes place even at low erlotinib concentrations. In the clinical setting, this interaction may result in increased exposure to erlotinib, especially in undernourished cancer patients. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Alejandro Pérez-Pitarch
- Pharmacy and Pharmaceutical Technology Department, University of Valencia, Valencia, Spain.,Pharmacy Department, University Clinical Hospital of Valencia, Valencia, Spain
| | - Beatriz Guglieri-López
- Pharmacy and Pharmaceutical Technology Department, University of Valencia, Valencia, Spain
| | - Amparo Nacher
- Pharmacy and Pharmaceutical Technology Department, University of Valencia, Valencia, Spain
| | - Virginia Merino
- Pharmacy and Pharmaceutical Technology Department, University of Valencia, Valencia, Spain
| | - Matilde Merino-Sanjuan
- Pharmacy and Pharmaceutical Technology Department, University of Valencia, Valencia, Spain
| |
Collapse
|
18
|
Peters SA, Jones CR, Ungell AL, Hatley OJD. Predicting Drug Extraction in the Human Gut Wall: Assessing Contributions from Drug Metabolizing Enzymes and Transporter Proteins using Preclinical Models. Clin Pharmacokinet 2016; 55:673-96. [PMID: 26895020 PMCID: PMC4875961 DOI: 10.1007/s40262-015-0351-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Intestinal metabolism can limit oral bioavailability of drugs and increase the risk of drug interactions. It is therefore important to be able to predict and quantify it in drug discovery and early development. In recent years, a plethora of models-in vivo, in situ and in vitro-have been discussed in the literature. The primary objective of this review is to summarize the current knowledge in the quantitative prediction of gut-wall metabolism. As well as discussing the successes of current models for intestinal metabolism, the challenges in the establishment of good preclinical models are highlighted, including species differences in the isoforms; regional abundances and activities of drug metabolizing enzymes; the interplay of enzyme-transporter proteins; and lack of knowledge on enzyme abundances and availability of empirical scaling factors. Due to its broad specificity and high abundance in the intestine, CYP3A is the enzyme that is frequently implicated in human gut metabolism and is therefore the major focus of this review. A strategy to assess the impact of gut wall metabolism on oral bioavailability during drug discovery and early development phases is presented. Current gaps in the mechanistic understanding and the prediction of gut metabolism are highlighted, with suggestions on how they can be overcome in the future.
Collapse
Affiliation(s)
- Sheila Annie Peters
- Translational Quantitative Pharmacology, BioPharma, R&D Global Early Development, Merck KGaA, Frankfurter Str. 250, F130/005, 64293, Darmstadt, Germany.
| | | | - Anna-Lena Ungell
- Investigative ADME, Non-Clinical Development, UCB New Medicines, BioPharma SPRL, Braine l'Alleud, Belgium
| | - Oliver J D Hatley
- Simcyp Limited (A Certara Company), Blades Enterprise Centre, Sheffield, UK
| |
Collapse
|
19
|
Song CK, Yoon IS, Kim DD. Poloxamer-based solid dispersions for oral delivery of docetaxel: Differential effects of F68 and P85 on oral docetaxel bioavailability. Int J Pharm 2016; 507:102-8. [PMID: 27154250 DOI: 10.1016/j.ijpharm.2016.05.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/11/2016] [Accepted: 05/02/2016] [Indexed: 10/21/2022]
Abstract
Development of an oral docetaxel formulation has been hindered mainly due to its poor solubility and oral bioavailability. The aim of this study was to develop poloxamer F68/P85-based solid dispersions (SDs) for the oral delivery of docetaxel and investigate their in vivo pharmacokinetic impacts on the systemic absorption of docetaxel given orally, in comparison with a SD based on F68 alone. The F68 and/or P85-based docetaxel SDs were prepared with varying the contents of poloxamers and then evaluated in terms of morphology, crystallinity, solubility, dissolution, permeation across rat intestinal segments, and oral pharmacokinetics in rats. As a result, the SDs successfully changed the crystalline properties of docetaxel and enhanced the drug solubility and dissolution. The SD prepared with F68 alone significantly enhanced the dissolution but not intestinal permeation of docetaxel, leading to only limited enhancement of oral bioavailability (1.39-fold increase). Notably, however, the F68/P85-based SD significantly enhanced both the dissolution and intestinal permeation of docetaxel, achieving a markedly improved oral bioavailability (2.97-fold increase). Therefore, the present results suggest that the intestinal permeation factor should be taken into account when designing SD formulations for the oral delivery of BCS class IV drugs including docetaxel, and that P85 could serve as a potential formulation excipient for enhancing the intestinal permeation of docetaxel.
Collapse
Affiliation(s)
- Chung Kil Song
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea; Institute of Nanoscience and Technology, Hanyang University, Seoul 04763, Republic of Korea
| | - In-Soo Yoon
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, 1666 Youngsan-ro, Muan-gun, Jeonnam 58554, Republic of Korea.
| | - Dae-Duk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
20
|
Vanhove T, Annaert P, Kuypers DRJ. Clinical determinants of calcineurin inhibitor disposition: a mechanistic review. Drug Metab Rev 2016; 48:88-112. [DOI: 10.3109/03602532.2016.1151037] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
21
|
Lo YL, Liu Y. Reversing multidrug resistance in Caco-2 by silencing MDR1, MRP1, MRP2, and BCL-2/BCL-xL using liposomal antisense oligonucleotides. PLoS One 2014; 9:e90180. [PMID: 24637737 PMCID: PMC3956467 DOI: 10.1371/journal.pone.0090180] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 01/27/2014] [Indexed: 12/16/2022] Open
Abstract
Multidrug resistance (MDR) is a major impediment to chemotherapy. In the present study, we designed antisense oligonucleotides (ASOs) against MDR1, MDR-associated protein (MRP)1, MRP2, and/or BCL-2/BCL-xL to reverse MDR transporters and induce apoptosis, respectively. The cationic liposomes (100 nm) composed of N-[1-(2,3-dioleyloxy)propyl]-n,n,n-trimethylammonium chloride and dioleoyl phosphotidylethanolamine core surrounded by a polyethylene glycol (PEG) shell were prepared to carry ASOs and/or epirubicin, an antineoplastic agent. We aimed to simultaneously suppress efflux pumps, provoke apoptosis, and enhance the chemosensitivity of human colon adenocarcinoma Caco-2 cells to epirubicin. We evaluated encapsulation efficiency, particle size, cytotoxicity, intracellular accumulation, mRNA levels, cell cycle distribution, and caspase activity of these formulations. We found that PEGylated liposomal ASOs significantly reduced Caco-2 cell viability and thus intensified epirubicin-mediated apoptosis. These formulations also decreased the MDR1 promoter activity levels and enhanced the intracellular retention of epirubicin in Caco-2 cells. Epirubicin and ASOs in PEGylated liposomes remarkably decreased mRNA expression levels of human MDR1, MRP1, MRP2, and BCL-2. The combined treatments all significantly increased the mRNA expressions of p53 and BAX, and activity levels of caspase-3, -8, and -9. The formulation of epirubicin and ASOs targeting both pump resistance of MDR1, MRP1, and MRP2 and nonpump resistance of BCL-2/BCL-xL demonstrated more superior effect to all the other formulations used in this study. Our results provide a novel insight into the mechanisms by which PEGylated liposomal ASOs against both resistance types act as activators to epirubicin-induced apoptosis through suppressing MDR1, MRP1, and MRP2, as well as triggering intrinsic mitochondrial and extrinsic death receptor pathways. The complicated regulation of MDR highlights the necessity for a multifunctional approach using an effective delivery system, such as PEGylated liposomes, to carry epirubicin and ASOs as a potent nanomedicine for improving the clinical efficacy of chemotherapy.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- Apoptosis/drug effects
- Apoptosis/genetics
- Caco-2 Cells
- Caspases/metabolism
- Cell Cycle/drug effects
- DNA Fragmentation/drug effects
- Drug Resistance, Neoplasm/genetics
- Epirubicin/administration & dosage
- Gene Expression
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Silencing
- Genes, Reporter
- Humans
- Liposomes
- Multidrug Resistance-Associated Protein 2
- Multidrug Resistance-Associated Proteins/genetics
- Oligonucleotides, Antisense/administration & dosage
- Oligonucleotides, Antisense/chemistry
- Oligonucleotides, Antisense/toxicity
- Polyethylene Glycols
- Promoter Regions, Genetic
- Proto-Oncogene Proteins c-bcl-2/genetics
- RNA, Messenger/genetics
- bcl-X Protein/genetics
Collapse
Affiliation(s)
- Yu-Li Lo
- Department of Biological Sciences and Technology, National University of Tainan, Tainan, Taiwan
- * E-mail:
| | - Yu Liu
- Department of Biological Sciences and Technology, National University of Tainan, Tainan, Taiwan
| |
Collapse
|
22
|
Werle M, Takeuchi H, Bernkop-Schnürch A. New-generation efflux pump inhibitors. Expert Rev Clin Pharmacol 2014; 1:429-40. [DOI: 10.1586/17512433.1.3.429] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
23
|
Choi YH, Yu AM. ABC transporters in multidrug resistance and pharmacokinetics, and strategies for drug development. Curr Pharm Des 2014; 20:793-807. [PMID: 23688078 PMCID: PMC6341993 DOI: 10.2174/138161282005140214165212] [Citation(s) in RCA: 430] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Accepted: 05/09/2013] [Indexed: 12/18/2022]
Abstract
Multidrug resistance (MDR) is a serious problem that hampers the success of cancer pharmacotherapy. A common mechanism is the overexpression of ATP-binding cassette (ABC) efflux transporters in cancer cells such as P-glycoprotein (P-gp/ABCB1), multidrug resistance-associated protein 1 (MRP1/ABCC1) and breast cancer resistance protein (BCRP/ABCG2) that limit the exposure to anticancer drugs. One way to overcome MDR is to develop ABC efflux transporter inhibitors to sensitize cancer cells to chemotherapeutic drugs. The complete clinical trials thus far have showen that those tested chemosensitizers only add limited or no benefits to cancer patients. Some MDR modulators are merely toxic, and others induce unwanted drug-drug interactions. Actually, many ABC transporters are also expressed abundantly in the gastrointestinal tract, liver, kidney, brain and other normal tissues, and they largely determine drug absorption, distribution and excretion, and affect the overall pharmacokinetic properties of drugs in humans. In addition, ABC transporters such as P-gp, MRP1 and BCRP co-expressed in tumors show a broad and overlapped specificity for substrates and MDR modulators. Thus reliable preclinical assays and models are required for the assessment of transporter-mediated flux and potential effects on pharmacokinetics in drug development. In this review, we provide an overview of the role of ABC efflux transporters in MDR and pharmacokinetics. Preclinical assays for the assessment of drug transport and development of MDR modulators are also discussed.
Collapse
Affiliation(s)
| | - Ai-Ming Yu
- Biochemistry & Molecular Medicine, UC Davis Medical Center, 2700 Stockton Blvd., Suite 2132, Sacramento, CA 95817, USA.
| |
Collapse
|
24
|
Estudante M, Maya M, Morais JG, Soveral G, Benet LZ. Effect of P-glycoprotein on the rat intestinal permeability and metabolism of the BDDCS class 1 drug verapamil. Mol Pharm 2013; 10:4038-45. [PMID: 24044638 DOI: 10.1021/mp400381p] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The Biopharmaceutics Drug Disposition Classification System (BDDCS) predicts intestinal transporter effects to be clinically insignificant following oral dosing for highly soluble and highly permeable/metabolized drugs (class 1 drugs). We investigated the effect of inhibiting P-glycoprotein (P-gp) on the in vitro rat intestinal permeability (Papp) and metabolism of the class 1 drug verapamil. Jejunal segments from Sprague-Dawley rats fasted overnight were mounted in Ussing chambers filled with 10 mL of Krebs-Ringer buffer (KRB). For P-gp inhibition studies, GG918 0.5 μM was added to the KRB solution. The experiment started by the addition of verapamil (1 or 10 μM) to either apical or basolateral sides. Samples from verapamil donor and receiver compartments were collected at 30 s and 0.166, 0.5, 1, 1.83 and 3 h after the start of the experiment. Analysis of verapamil and its major metabolite, norverapamil, in the samples and intracellularly at 3 h was performed by HPLC. The same experiment was repeated with norverapamil 10 μM (verapamil metabolite), digoxin 100 nM (positive control for P-gp activity) and atorvastatin 1 and 10 μM (example of a class 2 drug). For 1 μM verapamil, efflux ratio (B to A Papp/A to B Papp) was 4.6 and markedly decreased by GG918 (efflux ratio = 1.1). For 10 μM verapamil efflux ratio was 4.1 (control) vs 1.8 (GG918), comparable to the change seen for digoxin 100 nM with an efflux ratio of 3.6 (control) vs 1.6 (with GG918) and atorvastatin (efflux ratio of 5.2 and 3.0 for atorvastatin 1.0 and 10 μM, respectively, changed to 1.0 and 0.65 with GG918). The changes observed in the norverapamil 10 μM experiment were also significant, where efflux ratio decreased from 13.5 (control) to 1.5 (GG918). The extraction ratio (ER) of 10 μM verapamil to norverapamil decreased from 0.41 after an apical dose to 0.21 after a basolateral dose, but was unaffected by the incubation with GG918. The results suggest that P-gp inhibition has an effect on class 1 drug verapamil and class 2 drug atorvastatin Papp in the rat intestine. Moreover, a stronger P-gp effect on the Papp of the more polar norverapamil metabolite was observed. Papp changes caused by the P-gp inhibitor GG918 do not affect the extent of verapamil metabolism.
Collapse
Affiliation(s)
- Margarida Estudante
- iMed.UL, Faculty of Pharmacy, University of Lisbon , Av. Prof. Gama Pinto, 1649-019 Lisbon, Portugal
| | | | | | | | | |
Collapse
|
25
|
Thanki K, Gangwal RP, Sangamwar AT, Jain S. Oral delivery of anticancer drugs: Challenges and opportunities. J Control Release 2013; 170:15-40. [DOI: 10.1016/j.jconrel.2013.04.020] [Citation(s) in RCA: 328] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 04/25/2013] [Accepted: 04/26/2013] [Indexed: 12/12/2022]
|
26
|
Yano K, Tomono T, Sakai R, Kano T, Morimoto K, Kato Y, Ogihara T. Contribution of Radixin to P-Glycoprotein Expression and Transport Activity in Mouse Small Intestine In Vivo. J Pharm Sci 2013; 102:2875-81. [DOI: 10.1002/jps.23637] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 05/14/2013] [Accepted: 05/21/2013] [Indexed: 11/11/2022]
|
27
|
Senanayake TH, Warren G, Wei X, Vinogradov SV. Application of activated nucleoside analogs for the treatment of drug-resistant tumors by oral delivery of nanogel-drug conjugates. J Control Release 2013; 167:200-9. [PMID: 23385032 DOI: 10.1016/j.jconrel.2013.01.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 01/18/2013] [Accepted: 01/26/2013] [Indexed: 01/20/2023]
Abstract
A majority of nanoencapsulated drugs that have shown promise in cancer chemotherapy are administered intravenously. Development of effective oral nanoformulations presents a very challenging medical goal. Here, we describe successful applications of innovative polymeric nanogels in the form of conjugates with activated nucleoside analogs for oral administration in cancer chemotherapy. Previously, we reported the synthesis of amphiphilic polyvinyl alcohol and dextrin-based nanogel conjugates with the phosphorylated 5-FU nucleoside Floxuridine and demonstrated their enhanced activity against regular and drug-resistant cancers (T.H. Senanayake, G. Warren, S.V. Vinogradov, Novel anticancer polymeric conjugates of activated nucleoside analogs, Bioconjug. Chem. 22 (2011) 1983-1993). In this study, we synthesized and evaluated oral applications of nanogel conjugates of a protected Gemcitabine, the drug never used in oral therapies. These conjugates were able to quickly release an active form of the drug (Gemcitabine 5'-mono-, di- and triphosphates) by specific enzymatic activities, or slowly during hydrolysis. Gemcitabine conjugates demonstrated up to 127 times higher in vitro efficacy than the free drug against various cancer cells, including the lines resistant to nucleoside analogs. Surprisingly, these nanogel-drug conjugates were relatively stable in gastric conditions and able to actively penetrate through the gastrointestinal barrier based on permeability studies in Caco-2 cell model. In tumor xenograft models of several drug-resistant human cancers, we observed an efficient inhibition of tumor growth and extended the life-span of the animals by 3 times that of the control with orally treated Gemcitabine- or Floxuridine-nanogel conjugates. Thus, we have demonstrated a potential of therapeutic nanogel conjugates with the activated and stabilized Gemcitabine as a successful oral drug form against Gemcitabine-resistant and other drug-resistant tumors.
Collapse
Affiliation(s)
- Thulani H Senanayake
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, Omaha, United States
| | | | | | | |
Collapse
|
28
|
Comprehensive study on regional human intestinal permeability and prediction of fraction absorbed of drugs using the Ussing chamber technique. Eur J Pharm Sci 2013; 48:166-80. [DOI: 10.1016/j.ejps.2012.10.007] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 10/12/2012] [Accepted: 10/13/2012] [Indexed: 01/21/2023]
|
29
|
Wada S, Kano T, Mita S, Idota Y, Morimoto K, Yamashita F, Ogihara T. The Role of Inter-segmental Differences in P-glycoprotein Expression and Activity along the Rat Small Intestine in Causing the Double-peak Phenomenon of Substrate Plasma Concentration. Drug Metab Pharmacokinet 2013; 28:98-103. [DOI: 10.2133/dmpk.dmpk-12-rg-005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
30
|
Oga EF, Sekine S, Horie T. Ex Vivo and In Vivo Investigations of the Effects of Extracts of Vernonia amygdalina, Carica papaya and Tapinanthus sessilifolius on Digoxin Transport and Pharmacokinetics: Assessing the Significance on Rat Intestinal P-glycoprotein Efflux. Drug Metab Pharmacokinet 2013; 28:314-20. [DOI: 10.2133/dmpk.dmpk-12-rg-117] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
31
|
YANG SIHYUNG, CHOI HYEONGYEOM, LIM SOOJEONG, LEE MYUNGGULL, KIM SOHEE. Effects of morin on the pharmacokinetics of etoposide in 7,12-dimethylbenz[a]anthracene-induced mammary tumors in female Sprague-Dawley rats. Oncol Rep 2012; 29:1215-23. [DOI: 10.3892/or.2012.2201] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 11/22/2012] [Indexed: 11/05/2022] Open
|
32
|
Harwood MD, Neuhoff S, Carlson GL, Warhurst G, Rostami-Hodjegan A. Absolute abundance and function of intestinal drug transporters: a prerequisite for fully mechanisticin vitro-in vivoextrapolation of oral drug absorption. Biopharm Drug Dispos 2012; 34:2-28. [DOI: 10.1002/bdd.1810] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 08/01/2012] [Accepted: 08/13/2012] [Indexed: 12/14/2022]
Affiliation(s)
| | - S. Neuhoff
- Simcyp Ltd (a Certara Company); Blades Enterprise Centre; Sheffield; S2 4SU; UK
| | - G. L. Carlson
- Gut Barrier Group, School of Translational Medicine; University of Manchester, Salford Royal Hospital NHS Trust; M6 8HD; UK
| | - G. Warhurst
- Gut Barrier Group, School of Translational Medicine; University of Manchester, Salford Royal Hospital NHS Trust; M6 8HD; UK
| | | |
Collapse
|
33
|
Mandal M, Zhu Z, Cumming JN, Liu X, Strickland C, Mazzola RD, Caldwell JP, Leach P, Grzelak M, Hyde L, Zhang Q, Terracina G, Zhang L, Chen X, Kuvelkar R, Kennedy ME, Favreau L, Cox K, Orth P, Buevich A, Voigt J, Wang H, Kazakevich I, McKittrick BA, Greenlee W, Parker EM, Stamford AW. Design and Validation of Bicyclic Iminopyrimidinones As Beta Amyloid Cleaving Enzyme-1 (BACE1) Inhibitors: Conformational Constraint to Favor a Bioactive Conformation. J Med Chem 2012; 55:9331-45. [DOI: 10.1021/jm301039c] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Mihirbaran Mandal
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Zhaoning Zhu
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jared N. Cumming
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xiaoxiang Liu
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Corey Strickland
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Robert D. Mazzola
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - John P. Caldwell
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Prescott Leach
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Michael Grzelak
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lynn Hyde
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Qi Zhang
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Giuseppe Terracina
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lili Zhang
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xia Chen
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Reshma Kuvelkar
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Matthew E. Kennedy
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Leonard Favreau
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Kathleen Cox
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Peter Orth
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Alexei Buevich
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Johannes Voigt
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Hongwu Wang
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Irina Kazakevich
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Brian A. McKittrick
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - William Greenlee
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Eric M. Parker
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Andrew W. Stamford
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| |
Collapse
|
34
|
Jin HE, Song B, Kim SB, Shim WS, Kim DD, Chong S, Chung SJ, Shim CK. Transport of gemifloxacin, a 4th generation quinolone antibiotic, in the Caco-2 and engineered MDCKII cells, and potential involvement of efflux transporters in the intestinal absorption of the drug. Xenobiotica 2012; 43:355-67. [DOI: 10.3109/00498254.2012.720740] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
35
|
Sun ZY, Asberom T, Bara T, Bennett C, Burnett D, Chu I, Clader J, Cohen-Williams M, Cole D, Czarniecki M, Durkin J, Gallo G, Greenlee W, Josien H, Huang X, Hyde L, Jones N, Kazakevich I, Li H, Liu X, Lee J, MacCoss M, Mandal MB, McCracken T, Nomeir A, Mazzola R, Palani A, Parker EM, Pissarnitski DA, Qin J, Song L, Terracina G, Vicarel M, Voigt J, Xu R, Zhang L, Zhang Q, Zhao Z, Zhu X, Zhu Z. Cyclic Hydroxyamidines as Amide Isosteres: Discovery of Oxadiazolines and Oxadiazines as Potent and Highly Efficacious γ-Secretase Modulators in Vivo. J Med Chem 2011; 55:489-502. [DOI: 10.1021/jm201407j] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Zhong-Yue Sun
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Theodros Asberom
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Thomas Bara
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Chad Bennett
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Duane Burnett
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Inhou Chu
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - John Clader
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Mary Cohen-Williams
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - David Cole
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Michael Czarniecki
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - James Durkin
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Gioconda Gallo
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - William Greenlee
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Hubert Josien
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xianhai Huang
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lynn Hyde
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Nicholas Jones
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Irina Kazakevich
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Hongmei Li
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xiaoxiang Liu
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Julie Lee
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Malcolm MacCoss
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Mihir B. Mandal
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Troy McCracken
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Amin Nomeir
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Robert Mazzola
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Anandan Palani
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Eric M. Parker
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Dmitri A. Pissarnitski
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jun Qin
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lixin Song
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Giuseppe Terracina
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Monica Vicarel
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Johannes Voigt
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Ruo Xu
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lili Zhang
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Qi Zhang
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Zhiqiang Zhao
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xiaohong Zhu
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Zhaoning Zhu
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| |
Collapse
|
36
|
Watson KJ, Davis J, Jones HM. Application of physiologically based pharmacokinetic modeling to understanding the clinical pharmacokinetics of UK-369,003. Drug Metab Dispos 2011; 39:1203-13. [PMID: 21451120 DOI: 10.1124/dmd.111.038224] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
5-[2-Ethoxy-5-(4-ethyl-piperazine-1-sulfonyl)-pyridin-3-yl]-3-ethyl-2-(2-methoxy-ethyl)-2,6-dihydro-pyrazolo[4,3-d]pyrimidin-7-one (UK-369,003) is a phosphodiesterase-5 inhibitor in clinical development at Pfizer. UK-369,003 is predominantly metabolized by cytochrome P450 3A4 and is also a substrate for the efflux transporter P-glycoprotein. The pharmacokinetics of UK-369,003 has been profiled after oral administration of 1 to 800 mg of an immediate release formulation to healthy volunteers. Nonlinearity was observed in the systemic exposure at doses of 100 mg and greater. In addition, the pharmacokinetics of UK-369,003 has also been investigated after oral administration of the more therapeutically attractive modified release formulation. Systemic exposure was prolonged with the modified release formulation, but bioavailability was reduced in comparison with that of the immediate release formulation. Physiologically based pharmacokinetic modeling strategies are commonly used in drug discovery and development. This work describes application of the physiologically based pharmacokinetic software GastroPlus to understand the pharmacokinetics of UK-369,003. The impact of gut wall and hepatically mediated CYP3A4 metabolism, in addition to the actions of P-glycoprotein, in causing the nonlinear pharmacokinetics of the immediate release formulation and the reduced bioavailability of the modified release form, was investigated. The model accurately described the systemic exposure of UK-369,003 after intravenous and both immediate and modified release oral administration and suggested that CYP3A4 is responsible for the majority of the nonlinearity in systemic exposure observed after administration of the immediate release form. Conversely, the reduced bioavailability of the modified release formulation is believed to be caused by incomplete release from the device, incomplete absorption of released drug, and, to a lesser extent, CYP3A4 metabolism.
Collapse
Affiliation(s)
- Kenny J Watson
- Department of Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide R&D, Ramsgate Road, Sandwich, Kent, CT13 9NJ, UK
| | | | | |
Collapse
|
37
|
Suh J, Kang H, Yoon I, Yang S, Kim S, Lee H, Shim CK, Lee M. Cysteine effects on the pharmacokinetics of etoposide in protein–calorie malnutrition rats: increased gastrointestinal absorption by cysteine. Xenobiotica 2011; 41:885-94. [DOI: 10.3109/00498254.2011.585186] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
38
|
Enhancing effect of N-octyl-O-sulfate chitosan on etoposide absorption. Int J Pharm 2011; 409:38-45. [DOI: 10.1016/j.ijpharm.2011.02.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 02/14/2011] [Accepted: 02/17/2011] [Indexed: 11/19/2022]
|
39
|
Lee CK, Ki SH, Choi JS. Effects of oral curcumin on the pharmacokinetics of intravenous and oral etoposide in rats: possible role of intestinal CYP3A and P-gp inhibition by curcumin. Biopharm Drug Dispos 2011; 32:245-51. [DOI: 10.1002/bdd.754] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
40
|
Najar I, Sharma S, Singh G, Koul S, Gupta P, Javed S, Johri R. Involvement of P-glycoprotein and CYP 3A4 in the enhancement of etoposide bioavailability by a piperine analogue. Chem Biol Interact 2011; 190:84-90. [DOI: 10.1016/j.cbi.2011.02.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Revised: 02/07/2011] [Accepted: 02/09/2011] [Indexed: 10/18/2022]
|
41
|
Zhang X, Qiu F, Jiang J, Gao C, Tan Y. Intestinal absorption mechanisms of berberine, palmatine, jateorhizine, and coptisine: involvement of P-glycoprotein. Xenobiotica 2011; 41:290-6. [PMID: 21319959 DOI: 10.3109/00498254.2010.529180] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The absorption and transport mechanisms of berberine, palmatine, jateorhizine, and coptisine were studied using a Caco-2 cells uptake and transport model, with the addition of cyclosporin A and verapamil as P-glycoprotein (P-gp) inhibitors and MK-571 as a multidrug resistance-associated protein 2 (MRP(2)) inhibitor. In the uptake experiment, berberine, palmatine, jateorhizine, and coptisine were all taken into Caco-2 cells, and their uptakes were increased in the presence of cyclosporin A or verapamil. In the transport experiment, P(app) (AP-BL) was between 0.1 and 1.0 × 10(6) cm/sec for berberine, palmatine, jateorhizine, and coptisine and was lower than P(app) (BL-AB). ER values were all >2. Cyclosporin A and verapamil both increased P(app) (AP-BL) but decreased P(app) (BL-AB) for berberine, palmatine, jateorhizine, and coptisine; ER values were decreased by >50%. MK-571 had no influence on the transmembrane transport of berberine, palmatine, jateorhizine, and coptisine. At a concentration of 1-100 μM, berberine, palmatine, jateorhizine, and coptisine had no significant effects on the bidirection transport of Rho123. Berberine, palmatine, jateorhizine, and coptisine were all P-gp substrates; and at the range of 1-100 μM, berberine, palmatine, jateorhizine, and coptisine had no inhibitory effects on P-gp.
Collapse
Affiliation(s)
- Xinfeng Zhang
- Department of Clinical Pharmacology, Shuguang Hospital Affiliated to Shanghai University of TCM, Shanghai, China
| | | | | | | | | |
Collapse
|
42
|
Press B. Optimization of the Caco-2 permeability assay to screen drug compounds for intestinal absorption and efflux. Methods Mol Biol 2011; 763:139-54. [PMID: 21874449 DOI: 10.1007/978-1-61779-191-8_9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
In vitro permeability assays are a valuable tool for scientists during lead compound optimization. As a majority of discovery projects are focused on the development of orally bioavailable drugs, correlation of in vitro permeability data to in vivo absorption results is critical for understanding the structural-physicochemical relationship (SPR) of drugs exhibiting low levels of absorption. For more than a decade, the Caco-2 screening assay has remained a popular, in vitro system to test compounds for both intestinal permeability and efflux liability. Despite advances in artificial membrane technology and in silico modeling systems, drug compounds still benefit from testing in cell-based epithelial monolayer assays for lead optimization. This chapter provides technical information for performing and optimizing the Caco-2 assay. In addition, techniques are discussed for dealing with some of the most pressing issues surrounding in vitro permeability assays (i.e., low aqueous solubility of test compounds and low postassay recovery). Insights are offered to help researchers avoid common pitfalls in the interpretation of in vitro permeability data, which can often lead to the perception of misleading results for correlation to in vivo data.
Collapse
Affiliation(s)
- Barry Press
- MaRS Centre, Ontario Institute for Cancer Research, Toronto, ON, Canada.
| |
Collapse
|
43
|
Hippalgaonkar K, Srirangam R, Avula B, Khan IA, Majumdar S. Interaction between topically and systemically coadministered P-glycoprotein substrates/inhibitors: effect on vitreal kinetics. Drug Metab Dispos 2010; 38:1790-7. [PMID: 20595378 PMCID: PMC2957161 DOI: 10.1124/dmd.110.032672] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 07/01/2010] [Indexed: 12/12/2022] Open
Abstract
The objective of the present study was to investigate the effect of topically coadministered P-glycoprotein (P-gp) substrates/inhibitors on the vitreal kinetics of a systemically administered P-gp substrate. Anesthetized male rabbits were used in these studies. The concentration-time profile of quinidine in the vitreous humor, after intravenous administration, was determined alone and in the presence of topically coadministered verapamil, prednisolone sodium phosphate (PP), and erythromycin. The vitreal pharmacokinetic parameters of quinidine in the presence of verapamil [apparent elimination rate constant (λ(z)), 0.0027 ± 0.0002 min(-1); clearance (CL_F), 131 ± 21 ml/min; area under the curve (AUC(0-∞)), 39 ± 7.0 μg · min/ml; and mean residence time, 435 ± 20 min] were significantly different from those of the control (0.0058 ± 0.0006 min(-1), 296 ± 46 ml/min, 17 ± 3 μg · min/ml, and 232 ± 20 min, respectively). A 1.7-fold decrease in the vitreal λ(z) and a 1.5-fold increase in the vitreal AUC of quinidine were observed in the presence of topical PP. Statistically significant differences between the vitreal profiles of the control and erythromycin-treated group were also observed. Plasma concentration-time profiles of quinidine, alone or in the presence of the topically instilled compounds, remained unchanged, indicating uniform systemic quinidine exposure across groups. This study demonstrates an interaction between topically and systemically coadministered P-gp substrates, probably through the modulation of P-gp on the basolateral membrane of the retinal pigmented epithelium, leading to changes in the vitreal kinetics of the systemically administered agent.
Collapse
Affiliation(s)
- Ketan Hippalgaonkar
- Department of Pharmaceutics, School of Pharmacy, The University of Mississippi, University, MS 38677, USA
| | | | | | | | | |
Collapse
|
44
|
Kudela P, Koller VJ, Lubitz W. Bacterial ghosts (BGs)—Advanced antigen and drug delivery system. Vaccine 2010; 28:5760-7. [DOI: 10.1016/j.vaccine.2010.06.087] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 06/11/2010] [Accepted: 06/28/2010] [Indexed: 11/28/2022]
|
45
|
Mudra DR, Borchardt RT. Absorption Barriers in the Rat Intestinal Mucosa: 1. Application of an In Situ Perfusion Model to Simultaneously Assess Drug Permeation and Metabolism. J Pharm Sci 2010; 99:982-98. [DOI: 10.1002/jps.21912] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
46
|
Abstract
Delivery of biologically active agents to animals is often perceived to be the poor relation of human drug delivery. Yet this field has a long and successful history of species-specific device and formulation development, ranging from simple approaches and devices used in production animals to more sophisticated formulations and approaches for a wide range of species. While several technologies using biodegradable polymers have been successfully marketed in a range of veterinary and human products, the transfer of delivery technologies has not been similarly applied across species. This may be due to a combination of specific technical requirements for use of devices in different species, inter-species pharmacokinetic, pharmacodynamic and physiological differences, and distinct market drivers for drug classes used in companion and food-producing animals. This chapter reviews selected commercialised and research-based parenteral and non-parenteral veterinary drug delivery technologies in selected domestic species. Emphasis is also placed on the impact of endogenous drug transporters on drug distribution characteristics in different species. In vitro models used to investigate carrier-dependent transport are reviewed. Species-specific expression of transporters in several tissues can account for inter-animal or inter-species pharmacokinetic variability, lack of predictability of drug efficacy, and potential drug-drug interactions.
Collapse
Affiliation(s)
- David J Brayden
- School of Agriculture, Food Science and Veterinary Medicine, University College Dublin (UCD) and UCD Conway Institute, Belfield, Dublin 4, Ireland.
| | | | | |
Collapse
|
47
|
Petri N, Lennernäs H. In Vivo
Permeability Studies in the Gastrointestinal Tract of Humans. ACTA ACUST UNITED AC 2009. [DOI: 10.1002/9783527623860.ch9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
48
|
Bolger MB, Fraczkiewicz R, Lukacova V. Simulations of Absorption, Metabolism, and Bioavailability. ACTA ACUST UNITED AC 2009. [DOI: 10.1002/9783527623860.ch17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
49
|
Kamiyama E, Sugiyama D, Nakai D, Miura SI, Okazaki O. Culture period-dependent change of function and expression of ATP-binding cassette transporters in Caco-2 cells. Drug Metab Dispos 2009; 37:1956-62. [PMID: 19505989 DOI: 10.1124/dmd.109.027490] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The objective of this study was to determine an appropriate culture period to assess whether a compound of interest is transported by efflux transporters such as human multidrug resistance 1 (hMDR1), human multidrug resistance-associated protein 2 (hMRP2), and human breast cancer resistance protein (hBCRP) in Caco-2 cells. Caco-2 cells were cultured on a Transwell for 1 to 6 weeks. The expression of these transporters in the mRNA and protein levels was examined using a real-time polymerase chain reaction and Western blotting, respectively. Transcellular transport activities using digoxin, ochratoxin A, olmesartan, and estrone-3-sulfate were also examined. Except for digoxin, the permeability coefficient (P(app)) ratio of the three compounds at 2 weeks was the highest in the periods tested. The P(app) ratio of digoxin at 2 weeks was higher than that at 3 weeks. The temporal expression profile of each transporter in the mRNA level was similar to that in the protein level, and the functions of hMRP2 and hBCRP were roughly correlated with the expression in the mRNA and protein levels, but that of hMDR1 was not. These data suggest that among all the culture periods evaluated a 2-week culture is the best culture period for transport studies to identify whether a compound is a substrate for hMDR1, hMRP2, and hBCRP.
Collapse
Affiliation(s)
- Emi Kamiyama
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan.
| | | | | | | | | |
Collapse
|
50
|
Bramuglia GF, Cortada CM, Curras V, Höcht C, Buontempo F, Mato G, Niselman V, Rubio M, Carballo M. Relationship between P-glycoprotein activity measured in peripheral blood mononuclear cells and indinavir bioavailability in healthy volunteers. J Pharm Sci 2009; 98:327-36. [PMID: 18506817 DOI: 10.1002/jps.21411] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Indinavir, a HIV-1 protease inhibitor, showed large inter-individual pharmacokinetic variability. It has been proposed as a substrate of P-glycoprotein (P-gp), an efflux transporter, that may contribute to limit indinavir bioavailability. A liquid formulation of indinavir was developed from indinavir capsules in order to study indinavir pharmacokinetics in healthy volunteers. Compartmental and noncompartmental analysis of indinavir plasma concentrations showed high inter-individual variability in terms of area under the curve (AUC) and maximal plasma concentration (C(max)). A significant negative association between AUC normalized to body weight (AUC x weight) and lymphocyte P-gp activity, using Rh123 efflux assay, was observed (p = 0.008; r = -0.75). AUC normalized to elimination rate constant (AUC x beta) also showed a significant negative relationship with lymphocyte P-gp activity (p = 0.03, r = -0.64). Apparent clearance (CL/[F x weight]) and volume of distribution (VD/[F x weight]) showed a positive correlation with P-gp activity. Conversely, elimination rate constant did not correlate with P-gp activity. Although there is not enough evidence of a correlation between lymphocitary and intestinal function of P-gp, our results suggest a relationship between a P-gp phenotype marker, Rh123 efflux assay in lymphocytes, and indinavir bioavailability.
Collapse
Affiliation(s)
- Guillermo F Bramuglia
- Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, 5to piso (C1113AAD), Buenos Aires, Argentina.
| | | | | | | | | | | | | | | | | |
Collapse
|