1
|
Lu D, Rege B, Raw A, Yang J, Alam K, Bode C, Zhao L, Faustino P, Wu F, Shakleya D, Nickum E, Li BV, Wang R, Stier E, Miezeiewski B, Patel R, Boam A, Lionberger R, Keire D, Yu L. Antioxidants had No Effects on the In-Vitro Permeability of BCS III Model Drug Substances. J Pharm Sci 2024; 113:2708-2714. [PMID: 38862090 DOI: 10.1016/j.xphs.2024.05.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 06/13/2024]
Abstract
Reformulation with addition of antioxidants is one potential mitigation strategy to prevent or reduce nitrosamine drug substance-related impurities (NDSRIs) in drug products. To explore whether there could be other approaches to demonstrate bioequivalence for a reformulated oral product, which typically needs in vivo bioequivalence studies to support the changes after approval, the effects of antioxidant on the in vitro permeability of BCS III model drug substances were investigated to see whether there could be any potential impact on drug absorption. Six antioxidants were screened and four (ascorbic acid, cysteine, α-tocopherol and propyl gallate) were selected based on their nitrosamine inhibition efficiencies. The study demonstrated that these four antioxidants, at the tested amounts, did not have observable impact on the in vitro permeability of the BCS III model drug substances across Caco-2 cell monolayers in the In Vitro Dissolution Absorption System (IDAS). An in vitro permeability study could be considered as part of one potential bioequivalence bridging approach for reformulated low-risk immediate release solid oral products and oral suspension products. Other factors such as the influence of antioxidants on intestinal transporter activities should be considered where appropriate.
Collapse
Affiliation(s)
- Dongmei Lu
- Office of Pharmaceutical Quality, Center of Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA.
| | - Bhagwant Rege
- Office of Pharmaceutical Quality, Center of Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Andre Raw
- Office of Pharmaceutical Quality, Center of Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Jingyue Yang
- Office of Pharmaceutical Quality, Center of Drug Evaluation and Research, Food and Drug Administration, 645 Newstead Ave, Saint Louis, MO 63110, USA
| | - Khondoker Alam
- Office of Generic Drugs, Center of Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Chris Bode
- Pharmaron, 436 Creamery Way # 600G, Exton, PA 19341, USA
| | - Liang Zhao
- Office of Generic Drugs, Center of Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Patrick Faustino
- Office of Pharmaceutical Quality, Center of Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Fang Wu
- Office of Generic Drugs, Center of Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Diaa Shakleya
- Office of Pharmaceutical Quality, Center of Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Elisa Nickum
- Office of Pharmaceutical Quality, Center of Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Bing V Li
- Office of Generic Drugs, Center of Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Rong Wang
- Office of Generic Drugs, Center of Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Ethan Stier
- Office of Clinical Pharmacology, Office of Translational Sciences, Center of Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | | | - Rachana Patel
- Pharmaron, 436 Creamery Way # 600G, Exton, PA 19341, USA
| | - Ashley Boam
- Office of Pharmaceutical Quality, Center of Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Robert Lionberger
- Office of Generic Drugs, Center of Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - David Keire
- Office of Pharmaceutical Quality, Center of Drug Evaluation and Research, Food and Drug Administration, 645 Newstead Ave, Saint Louis, MO 63110, USA
| | - Lawrence Yu
- Office of Pharmaceutical Quality, Center of Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| |
Collapse
|
2
|
Das S, Giri L, Majumdar S. Interaction-Based Perspective for Designing Polymer Biomaterial: A Strategic Approach to the Chitosan-Glycerophosphate System. ACS Biomater Sci Eng 2024; 10:4359-4373. [PMID: 38842569 DOI: 10.1021/acsbiomaterials.4c00723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
The conventional approach for developing any polymeric biomaterial is to follow protocols available in the literature and/or perform trial-and-error runs without a scientific basis. Here, we propose an analysis of a complex overlay of molecular interactions between drugs and polymers that provides a strategic pathway for biomaterial development. First, this work provides an innovative interaction-based method for developing an ocular formulation involving in situ gelling chitosan, gelatin, and glycerophosphate systems. A systematic interaction study is conducted based on the measurement of hydrodynamic radius, zeta potential, and viscosity with the sequential addition of formulation components. The increase in the hydrodynamic radius of the polymer with the addition of drugs can be interpreted as better diffusion of the drug inside the charged polymer chains and vice versa. Based on the knowledge of these interactions, a formulation has been designed that shows better drug release results with extended and sustained release compared to literature protocols, hence accentuating the importance of this study. An in-depth analysis of interactions can lead to a better understanding of the system. Second, we demonstrate the development of two dual-drug biomaterial systems, i.e., an in situ gelling and a liquid formulation at ocular surface temperature from the same polymers, which can be used as an ocular antiglaucoma formulation. Prior knowledge of the interactions between the drug polymers can be used to design a better formulation. The demonstrated application of this interaction-based protocol development can be extended universally to any biomaterial. This would provide a comprehensive idea about the properties and interactions of polymers and drugs, which can also serve as a base/starting point for a new formulation/biomaterial development.
Collapse
Affiliation(s)
- Sougat Das
- Department of Chemical Engineering, Indian Institute of Technology Hyderabad Telangana, Hyderabad 502285, India
| | - Lopamudra Giri
- Department of Chemical Engineering, Indian Institute of Technology Hyderabad Telangana, Hyderabad 502285, India
| | - Saptarshi Majumdar
- Department of Chemical Engineering, Indian Institute of Technology Hyderabad Telangana, Hyderabad 502285, India
| |
Collapse
|
3
|
Tzanova MM, Larsen BS, Birolo R, Cignolini S, Tho I, Chierotti MR, Perissutti B, Scaglione S, Stein PC, Hiorth M, Di Cagno MP. Shifting the Focus from Dissolution to Permeation: Introducing the Meso-fluidic Chip for Permeability Assessment (MCPA). J Pharm Sci 2024; 113:1319-1329. [PMID: 38104888 DOI: 10.1016/j.xphs.2023.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/12/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023]
Abstract
In response to the growing ethical and environmental concerns associated with animal testing, numerous in vitro tools of varying complexity and biorelevance have been developed and adopted in pharmaceutical research and development. In this work, we present one of these tools, i.e., the Meso-fluidic Chip for Permeability Assessment (MCPA), for the first time. The MCPA combines an artificial barrier (PermeaPad®) with an organ-on-chip device (MIVO®) and real-time automated concentration measurements, to yield a sustainable, yet effortless method for permeation testing. The system offers three major physiological aspects, i.e., a biomimetic membrane, an optimal membrane interfacial area-to-donor-volume-ratio (A/V) and a physiological flow on the acceptor/basolateral side, which makes the MPCA an ideal candidate for mechanistic studies and excellent in vivo bioavailability predictions. We validated the method with a handful of assorted drug compounds in unstirred and stirred donor conditions, before exploring its applicability as a tool for dissolution/permeation testing on a BCS class III/I drug (pyrazinamide) crystalline adducts and BCS class II/IV (hydrocortisone) amorphous solid dispersions. The results were highly reproducible and clearly displayed the method's potential for evaluating the performance of enabling formulations, and possibly even predicting in vivo performance. We believe that, upon further development, the MCPA will serve as a useful in vitro tool that could push sustainability into pharmaceutics by refining, reducing and replacing animal testing in early-stage drug development.
Collapse
Affiliation(s)
- Martina M Tzanova
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Sem Saelands vei 3, 0371 Oslo, Norway
| | - Bjarke Strøm Larsen
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Sem Saelands vei 3, 0371 Oslo, Norway
| | - Rebecca Birolo
- Department of Chemistry and NIS centre, University of Torino, Via P. Giuria 7, 10125 Torino, Italy
| | - Sara Cignolini
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Alfonso Valerio, 6/1, 34127 Trieste, Italy
| | - Ingunn Tho
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Sem Saelands vei 3, 0371 Oslo, Norway
| | - Michele R Chierotti
- Department of Chemistry and NIS centre, University of Torino, Via P. Giuria 7, 10125 Torino, Italy
| | - Beatrice Perissutti
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via Alfonso Valerio, 6/1, 34127 Trieste, Italy
| | - Silvia Scaglione
- National Research Council (CNR) and React4life S.p.A., Genoa, Italy
| | - Paul C Stein
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, 5230 Odense, Denmark
| | - Marianne Hiorth
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Sem Saelands vei 3, 0371 Oslo, Norway
| | - Massimiliano Pio Di Cagno
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Sem Saelands vei 3, 0371 Oslo, Norway.
| |
Collapse
|
4
|
Kalsoom S, Rasool MF, Imran I, Saeed H, Ahmad T, Alqahtani F. A Comprehensive Physiologically Based Pharmacokinetic Model of Nadolol in Adults with Renal Disease and Pediatrics with Supraventricular Tachycardia. Pharmaceuticals (Basel) 2024; 17:265. [PMID: 38399480 PMCID: PMC10891759 DOI: 10.3390/ph17020265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/03/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
Nadolol is a long-acting non-selective β-adrenergic antagonist that helps treat angina and hypertension. The current study aimed to develop and validate the physiologically based pharmacokinetic model (PBPK) of nadolol in healthy adults, renal-compromised, and pediatric populations. A comprehensive PBPK model was established by utilizing a PK-Sim simulator. After establishing and validating the model in healthy adults, pathophysiological changes i.e., blood flow, hematocrit, and GFR that occur in renal failure were incorporated in the developed model, and the drug exposure was assessed through Box plots. The pediatric model was also developed and evaluated by considering the renal maturation process. The validation of the models was carried out by visual predictive checks, calculating predicted to observed (Rpre/obs) and the average fold error (AFE) of PK parameters i.e., the area under the concentration-time curve (AUC0-t), the maximum concentration in plasma (Cmax), and CL (clearance). The presented PBPK model successfully simulates the nadolol PK in healthy adults, renal-impaired, and pediatric populations, as the Rpre/obs values of all PK parameters fall within the acceptable range. The established PBPK model can be useful in nadolol dose optimization in patients with renal failure and children with supraventricular tachycardia.
Collapse
Affiliation(s)
- Samia Kalsoom
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan;
| | - Muhammad Fawad Rasool
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan;
| | - Imran Imran
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan;
| | - Hamid Saeed
- Section of Pharmaceutics, University College of Pharmacy, Allama Iqbal Campus, University of the Punjab, Lahore 54000, Pakistan;
| | - Tanveer Ahmad
- Institute for Advanced Biosciences (IAB), CNRS UMR5309, INSERM U1209, Grenoble Alpes University, 38700 La Tronche, France;
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
5
|
Kus M, Ibragimow I, Piotrowska-Kempisty H. Caco-2 Cell Line Standardization with Pharmaceutical Requirements and In Vitro Model Suitability for Permeability Assays. Pharmaceutics 2023; 15:2523. [PMID: 38004503 PMCID: PMC10674574 DOI: 10.3390/pharmaceutics15112523] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/19/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023] Open
Abstract
The Caco-2 cell line derived from human colon carcinoma is commonly used to assess the permeability of compounds in in vitro conditions. Due to the significant increase in permeability studies using the Caco-2 cell line in recent years, the need to standardize this biological model seems necessary. The pharmaceutical requirements define only the acceptance criteria for the validation of the Caco-2 cell line and do not specify the protocol for its implementation. Therefore, the aim of this study is to review the conditions for permeability studies across the Caco-2 monolayer reported in the available literature concerning validation guidelines. We summarized the main aspects affecting the validation process of the Caco-2 cell line, including the culture conditions, cytotoxicity, cell differentiation process, and monolayer transport conditions, and the main conclusions may be useful in developing individual methods for preparing the cell line for validation purposes and further permeability research.
Collapse
Affiliation(s)
- Marta Kus
- Department of Toxicology, Poznan University of Medical Sciences, 30 Dojazd St., 60-631 Poznan, Poland;
- Research and Development Department of Ethifarm, Ethifarm Manufacturing Plant, 9 Stefana Zeromskiego St., 60-544 Poznan, Poland;
| | - Izabela Ibragimow
- Research and Development Department of Ethifarm, Ethifarm Manufacturing Plant, 9 Stefana Zeromskiego St., 60-544 Poznan, Poland;
| | - Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences, 30 Dojazd St., 60-631 Poznan, Poland;
- Department of Basic and Preclinical Science, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, 7 Gagarina St., 87-100 Torun, Poland
| |
Collapse
|
6
|
Macedo MH, Torras N, García-Díaz M, Barrias C, Sarmento B, Martínez E. The shape of our gut: Dissecting its impact on drug absorption in a 3D bioprinted intestinal model. BIOMATERIALS ADVANCES 2023; 153:213564. [PMID: 37482042 DOI: 10.1016/j.bioadv.2023.213564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/13/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
The small intestine is a complex organ with a characteristic architecture and a major site for drug and nutrient absorption. The three-dimensional (3D) topography organized in finger-like protrusions called villi increases surface area remarkably, granting a more efficient absorption process. The intestinal mucosa, where this process occurs, is a multilayered and multicell-type tissue barrier. In vitro intestinal models are routinely used to study different physiological and pathological processes in the gut, including compound absorption. Still, standard models are typically two-dimensional (2D) and represent only the epithelial barrier, lacking the cues offered by the 3D architecture and the stromal components present in vivo, often leading to inaccurate results. In this work, we studied the impact of the 3D architecture of the gut on drug transport using a bioprinted 3D model of the intestinal mucosa containing both the epithelial and the stromal compartments. Human intestinal fibroblasts were embedded in a previously optimized hydrogel bioink, and enterocytes and goblet cells were seeded on top to mimic the intestinal mucosa. The embedded fibroblasts thrived inside the hydrogel, remodeling the surrounding extracellular matrix. The epithelial cells fully covered the hydrogel scaffolds and formed a uniform cell layer with barrier properties close to in vivo. In particular, the villus-like model revealed overall increased permeability compared to a flat counterpart composed by the same hydrogel and cells. In addition, the efflux activity of the P-glycoprotein (P-gp) transporter was significantly reduced in the villus-like scaffold compared to a flat model, and the genetic expression of other drugs transporters was, in general, more relevant in the villus-like model. Globally, this study corroborates that the presence of the 3D architecture promotes a more physiological differentiation of the epithelial barrier, providing more accurate data on drug absorbance measurements.
Collapse
Affiliation(s)
- Maria Helena Macedo
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Núria Torras
- IBEC - Institute for Bioengineering of Catalonia, BIST - The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - María García-Díaz
- IBEC - Institute for Bioengineering of Catalonia, BIST - The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Cristina Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Elena Martínez
- IBEC - Institute for Bioengineering of Catalonia, BIST - The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain; CIBER-BBN - Consorcio Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Avenida Monforte de Lemos 3-5, 28029 Madrid, Spain; Electronics and Biomedical Engineering Department, Universitat de Barcelona, Martí I Franquès 1, 08028 Barcelona, Spain.
| |
Collapse
|
7
|
Habiballah S, Reisfeld B. Adapting physiologically-based pharmacokinetic models for machine learning applications. Sci Rep 2023; 13:14934. [PMID: 37696914 PMCID: PMC10495394 DOI: 10.1038/s41598-023-42165-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 09/06/2023] [Indexed: 09/13/2023] Open
Abstract
Both machine learning and physiologically-based pharmacokinetic models are becoming essential components of the drug development process. Integrating the predictive capabilities of physiologically-based pharmacokinetic (PBPK) models within machine learning (ML) pipelines could offer significant benefits in improving the accuracy and scope of drug screening and evaluation procedures. Here, we describe the development and testing of a self-contained machine learning module capable of faithfully recapitulating summary pharmacokinetic (PK) parameters produced by a full PBPK model, given a set of input drug-specific and regimen-specific information. Because of its widespread use in characterizing the disposition of orally administered drugs, the PBPK model chosen to demonstrate the methodology was an open-source implementation of a state-of-the-art compartmental and transit model called OpenCAT. The model was tested for drug formulations spanning a large range of solubility and absorption characteristics, and was evaluated for concordance against predictions of OpenCAT and relevant experimental data. In general, the values predicted by the ML models were within 20% of those of the PBPK model across the range of drug and formulation properties. However, summary PK parameter predictions from both the ML model and full PBPK model were occasionally poor with respect to those derived from experiments, suggesting deficiencies in the underlying PBPK model.
Collapse
Affiliation(s)
- Sohaib Habiballah
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO, 80523-1301, USA
| | - Brad Reisfeld
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO, 80523-1301, USA.
- School of Public Health, Colorado State University, Fort Collins, CO, 80523-1612, USA.
| |
Collapse
|
8
|
Uner B, Ozdemir S, Nur Pilevne S, Rıza Cenk Celebi A. Timolol-loaded ethosomes for ophthalmic delivery: Reduction of high intraocular pressure in vivo. Int J Pharm 2023; 640:123021. [PMID: 37149109 DOI: 10.1016/j.ijpharm.2023.123021] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/14/2023] [Accepted: 05/01/2023] [Indexed: 05/08/2023]
Abstract
The beta-adrenoceptor blocker timolol maleate (TML) is a commonly used pharmaceutical agent for the management of glaucoma. Conventional eye drops have limitations due to biological or pharmaceutical factors. Therefore, TML-loaded ethosomes have been designed to mitigate these restrictions and give a viable solution for reducing elevated intraocular pressure (IOP). The ethosomes were prepared using the thin film hydration method. Integrating the Box-Behnken experimental strategy, the optimal formulation was identified. The physicochemical characterization studies were performed on the optimal formulation. Then, in vitro release and ex vivo permeation studies were conducted. The irritation assessment was also carried out with Hen's Egg Test-Chorioallantoic Membrane model (HET-CAM), and in vivo evaluation of the IOP lowering effect was also performed on rats. The physicochemical characterization studies demonstrated that the components of the formulation were compatible with each other. The particle size, zeta potential, and encapsulation efficiency (EE%) were found as 88.23 ± 1.25 nm, -28.7 ± 2.03 mV, and 89.73 ± 0.42 %, respectively. The in vitro drug release mechanism was found as Korsmeyer-Peppas kinetics (R2=0.9923). The HET-CAM findings verified the formulation's eligibility for biological applications. The IOP measurements revealed no statistical difference (p>0.05) between the once-a-day application of the optimal formulation and the three-times-a-day application of the conventional eye drop. A similar pharmacological response was observed at lowered application frequencies. Therefore, it was concluded that the novel TML-loaded ethosomes could be a safe and efficient alternative for glaucoma treatment.
Collapse
Affiliation(s)
- Burcu Uner
- Yeditepe University, Faculty of Pharmacy, Department of Pharmaceutical Technology, Istanbul, Turkey; University of Health Sciences and Pharmacy in St. Louis, Department of Pharmaceutical and Administrative Sciences, St. Louis, MO, USA
| | - Samet Ozdemir
- Istanbul Health and Technology University, Faculty of Pharmacy, Department of Pharmaceutical Technology, Istanbul, Turkey.
| | - Seniz Nur Pilevne
- Acibadem Mehmet Ali Aydinlar University, School of Medicine, Department of Ophthalmology, Istanbul, Turkey
| | - Ali Rıza Cenk Celebi
- Acibadem Mehmet Ali Aydinlar University, School of Medicine, Department of Ophthalmology, Istanbul, Turkey
| |
Collapse
|
9
|
Sahoo DK, Martinez MN, Dao K, Gabriel V, Zdyrski C, Jergens AE, Atherly T, Iennarella-Servantez CA, Burns LE, Schrunk D, Volpe DA, Allenspach K, Mochel JP. Canine Intestinal Organoids as a Novel In Vitro Model of Intestinal Drug Permeability: A Proof-of-Concept Study. Cells 2023; 12:cells12091269. [PMID: 37174669 PMCID: PMC10177590 DOI: 10.3390/cells12091269] [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: 03/31/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
A key component of efforts to identify the biological and drug-specific aspects contributing to therapeutic failure or unexpected exposure-associated toxicity is the study of drug-intestinal barrier interactions. While methods supporting such assessments are widely described for human therapeutics, relatively little information is available for similar evaluations in support of veterinary pharmaceuticals. There is, therefore, a critical need to develop novel approaches for evaluating drug-gut interactions in veterinary medicine. Three-dimensional (3D) organoids can address these difficulties in a reasonably affordable system that circumvents the need for more invasive in vivo assays in live animals. However, a first step in developing such systems is understanding organoid interactions in a 2D monolayer. Given the importance of orally administered medications for meeting the therapeutic need of companion animals, we demonstrate growth conditions under which canine-colonoid-derived intestinal epithelial cells survive, mature, and differentiate into confluent cell systems with high monolayer integrity. We further examine the applicability of this canine-colonoid-derived 2D model to assess the permeability of three structurally diverse, passively absorbed β-blockers (e.g., propranolol, metoprolol, and atenolol). Both the absorptive and secretive apparent permeability (Papp) of these drugs at two different pH conditions were evaluated in canine-colonoid-derived monolayers and compared with that of Caco-2 cells. This proof-of-concept study provides promising preliminary results with regard to the utility of canine-derived organoid monolayers for species-specific assessments of therapeutic drug passive permeability.
Collapse
Affiliation(s)
- Dipak Kumar Sahoo
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Marilyn N Martinez
- Office of New Animal Drug Evaluation, Center for Veterinary Medicine, Food and Drug Administration, Rockville, MD 20852, USA
| | - Kimberly Dao
- 3D Health Solutions, Iowa State University, Ames, IA 50011, USA
| | - Vojtech Gabriel
- Department of Biomedical Sciences, SMART Pharmacology, Iowa State University, Ames, IA 50011, USA
| | - Christopher Zdyrski
- 3D Health Solutions, Iowa State University, Ames, IA 50011, USA
- Department of Biomedical Sciences, SMART Pharmacology, Iowa State University, Ames, IA 50011, USA
| | - Albert E Jergens
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Todd Atherly
- 3D Health Solutions, Iowa State University, Ames, IA 50011, USA
| | | | - Laura E Burns
- Veterinary Diagnostic Laboratory, Iowa State University, Ames, IA 50011, USA
| | - Dwayne Schrunk
- Veterinary Diagnostic Laboratory, Iowa State University, Ames, IA 50011, USA
| | - Donna A Volpe
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20852, USA
| | - Karin Allenspach
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA 50011, USA
- 3D Health Solutions, Iowa State University, Ames, IA 50011, USA
| | - Jonathan P Mochel
- 3D Health Solutions, Iowa State University, Ames, IA 50011, USA
- Department of Biomedical Sciences, SMART Pharmacology, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
10
|
Lee SJ, Joo SA, Kim H, Lee Y, Chung SJ, Chae YJ, Maeng HJ. Involvement of CYP3A4 and MDR1 in altered metabolism and transport of indinavir in 1,25(OH) 2D 3-treated Caco-2 cells. Eur J Pharm Sci 2023; 183:106396. [PMID: 36736464 DOI: 10.1016/j.ejps.2023.106396] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/02/2023] [Accepted: 01/30/2023] [Indexed: 02/02/2023]
Abstract
Altered drug concentrations may induce unexpected toxicity or treatment failure; thus, understanding the factors that alter the pharmacokinetic profiles of drugs is crucial for optimal disease treatment. Vitamin D receptor (VDR), a nuclear receptor, regulates the expression of cytochrome P450 3A4 (CYP3A4) and multidrug resistance protein 1 (MDR1), which are crucial determinants of drug pharmacokinetics. In this study, we investigated the effects of 1α,25-dihydroxyvitamin D3 [1,25(OH)2D3], a VDR ligand, on the metabolism, transport, and pharmacokinetics of indinavir, a dual substrate of CYP3A4 and MDR1. 1,25(OH)2D3 treatment for three days upregulated the expression levels of CYP3A4 and MDR1 in Caco-2 cells and consequently led to an increase in the level of a metabolite formed via CYP3A4 (indinavir M6) and the efflux ratio of indinavir in transport study. The increase in the metabolic reaction was also confirmed through a metabolism assay performed using the lysate of 1,25(OH)2D3-treated Caco-2 cells. In the Ussing chamber study conducted with the rat intestine, 1,25(OH)2D3 treatment did not alter the transport of indinavir into the basolateral side but increased indinavir M6 formation. Similarly, plasma levels of the metabolite increased in 1,25(OH)2D3-treated rats; however, systemic exposure to indinavir led to insignificant alterations. Considering the overlapping substrate specificities for CYP3A4 and MDR1 and their significant roles in drug pharmacokinetics, VDR may play an important role in drug interactions of CYP3A4 and MDR1 substrates for accessing more effective and safe disease treatments.
Collapse
Affiliation(s)
- Su-Jin Lee
- College of Pharmacy, Gachon University, Incheon 21936, Korea
| | - Seul-A Joo
- College of Pharmacy, Gachon University, Incheon 21936, Korea
| | - Heejeong Kim
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Yunjong Lee
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Suk-Jae Chung
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Yoon-Jee Chae
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Woosuk University, Wanju 55338, Korea.
| | - Han-Joo Maeng
- College of Pharmacy, Gachon University, Incheon 21936, Korea.
| |
Collapse
|
11
|
Muschong P, Jin L, Schejbal J, Mezler M, Weinheimer M. Improvement of Workflows and Assay Reproducibility by The Introduction of "Assay-Ready" Culturing of MDCK Cells for Transport Studies. Pharm Res 2023; 40:1259-1270. [PMID: 36977814 DOI: 10.1007/s11095-023-03490-4] [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: 10/19/2022] [Accepted: 02/23/2023] [Indexed: 03/30/2023]
Abstract
OBJECTIVE In previous studies, we established and validated three Madin Darby Canine Kidney MDCKII cell lines, recombinantly modified with zinc finger nuclease (ZFN) technology. Here, we investigated the applicability of seeding these three canine P-gp deficient MDCK_ZFN cell lines, directly from frozen cryopreserved stocks without previous cultivation for efflux transporter and permeability studies. This technique is referred to as "assay-ready" and allows for highly standardized conduction of cell-based assays and shorter cultivation cycles. METHODS To obtain a rapid fitness of the cells for that purpose, a very gentle freezing and thawing protocol was applied. Assay-ready MDCK_ZFN cells were tested in bi-directional transport studies and compared to their traditionally cultured counterparts. Long-term performance robustness, human effective intestinal permeability (Peff) predictability and batch to batch variability were assessed. RESULTS Efflux ratios (ER) and apparent permeability (Papp) results were highly comparable between assay-ready and standard cultured cell lines with R2 values of 0.96 or higher. Papp to Peff correlations obtained from passive permeability with non-transfected cells were comparable independent of the cultivation regime. Long-term evaluation revealed robust performance of assay-ready cells and reduced data variability of reference compounds in 75% of cases compared to standard cultured MDCK_ZFN cells. CONCLUSION Assay-ready methodology for handling MDCK_ZFN cells allows more flexibility in assay planning and reduces performance fluctuations in assays caused by cell aging. Therefore, the assay-ready principle has proven superior over conventional cultivation for MDCK_ZFN cells and is considered as a key technology to optimize processes with other cellular systems.
Collapse
Affiliation(s)
- Patricia Muschong
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co KG, Knollstr, 67061, Ludwigshafen, Germany
| | - Liang Jin
- Quantitative, Translational & ADME Sciences, AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, USA
| | - Jan Schejbal
- Quantitative, Translational & ADME Sciences, AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA, 01605, USA
| | - Mario Mezler
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co KG, Knollstr, 67061, Ludwigshafen, Germany
| | - Manuel Weinheimer
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co KG, Knollstr, 67061, Ludwigshafen, Germany.
| |
Collapse
|
12
|
Adhikari A, Seo PR, Polli JE. Dissolution-Hollow Fiber Membrane (D-HFM) System to Anticipate Biopharmaceutics Risk of Tablets and Capsules. J Pharm Sci 2023; 112:751-759. [PMID: 36202250 DOI: 10.1016/j.xphs.2022.09.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/29/2022] [Accepted: 09/29/2022] [Indexed: 02/18/2023]
Abstract
A dissolution-hollow fiber membrane (D-HFM) system with relatively high area/volume ratio was previously characterized and showed favorably high percent drug absorption. Also, it's in vitro permeation constant (Kp.Ç.) was close to in vivo human permeation constant (kp). The objective of the current study was to predict the in vivo human absorption profile and biopharmaceutic performance of five drug products using the D-HFM system. Four immediate-release (IR) and one extended-release (ER) solid oral dosage form were subjected to the D-HFM system. Tablets and capsule dissolution were also measured using USP apparatus II. Drug solutions were also subjected to D-HFM testing. Predicted and observed absorption profiles in D-HFM system showed close agreement for each solid oral dosage form. Levy-Polli plots from D-HFM system successfully predicted the four IR products to be low biopharmaceutic risk due to permeation rate limited or mixed dissolution/permeation rate limited absorption, and successfully predicted metoprolol ER product to be high biopharmaceutic risk due to dissolution rate limited absorption. These observations showed potential of the in vitro D-HFM system to be utilized in biopharmaceutics risk assessment of in vivo tablet and capsule performance.
Collapse
Affiliation(s)
- Asmita Adhikari
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| | - Paul R Seo
- Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - James E Polli
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA.
| |
Collapse
|
13
|
Pollard TD, Seoane-Viaño I, Ong JJ, Januskaite P, Awwad S, Orlu M, Bande MF, Basit AW, Goyanes A. Inkjet drug printing onto contact lenses: Deposition optimisation and non-invasive dose verification. Int J Pharm X 2022; 5:100150. [PMID: 36593987 PMCID: PMC9804110 DOI: 10.1016/j.ijpx.2022.100150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Inkjet printing has the potential to advance the treatment of eye diseases by printing drugs on demand onto contact lenses for localised delivery and personalised dosing, while near-infrared (NIR) spectroscopy can further be used as a quality control method for quantifying the drug but has yet to be demonstrated with contact lenses. In this study, a glaucoma therapy drug, timolol maleate, was successfully printed onto contact lenses using a modified commercial inkjet printer. The drug-loaded ink prepared for the printer was designed to match the properties of commercial ink, whilst having maximal drug loading and avoiding ocular inflammation. This setup demonstrated personalised drug dosing by printing multiple passes. Light transmittance was found to be unaffected by drug loading on the contact lens. A novel dissolution model was built, and in vitro dissolution studies showed drug release over at least 3 h, significantly longer than eye drops. NIR was used as an external validation method to accurately quantify the drug dose. Overall, the combination of inkjet printing and NIR represent a novel method for point-of-care personalisation and quantification of drug-loaded contact lenses.
Collapse
Affiliation(s)
- Thomas D. Pollard
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Iria Seoane-Viaño
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK,Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Paraquasil Group (GI-2109), Faculty of Pharmacy, and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela (USC), Santiago de Compostela 15782, Spain
| | - Jun Jie Ong
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Patricija Januskaite
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Sahar Awwad
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Mine Orlu
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Manuel F. Bande
- Department of Ophthalmology, University Hospital of Santiago de Compostela, Ramon Baltar S/N, Santiago de Compostela 15706, Spain
| | - Abdul W. Basit
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK,FabRx Ltd., Henwood House, Henwood, Ashford TN24 8DH, UK,Corresponding authors at: Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| | - Alvaro Goyanes
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK,FabRx Ltd., Henwood House, Henwood, Ashford TN24 8DH, UK,Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma Group (GI-1645), Facultad de Farmacia, iMATUS and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela (USC), Santiago de Compostela 15782, Spain,Corresponding authors at: Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| |
Collapse
|
14
|
Drug Physicochemical Properties and Capsule Fill Determine Extent of Premature Gastric Release from Enteric Capsules. Pharmaceutics 2022; 14:pharmaceutics14112505. [PMID: 36432696 PMCID: PMC9695824 DOI: 10.3390/pharmaceutics14112505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
Intrinsically, enteric capsule shells offer several advantages compared to coating of dosage forms with enteric polymers. We undertook a systematic investigation to elucidate capsule-fill parameters that may result in premature gastric drug release from Vcaps® Enteric capsules (Lonza CHI, Morristown, NJ, USA). Four model drugs with different ionization and solubility profiles were investigated: acetaminophen, ketoprofen, trimethoprim and atenolol. Different fill loads, diluents and drug-to-diluent ratios were explored. Enteric capsules were filled with drug or drug and diluent powder mix and underwent USP II dissolution testing using mini-vessels and paddles. Capsules were tested in pH 2 (0.01 M HCl) or pH 4.5 (3.2 × 10-5 M HCl) 200 mL acid media to simulate normal, fasted or hypochlorhydric gastric pH, respectively. Acetaminophen, trimethoprim and atenolol displayed premature gastric drug release from enteric capsules. The extent of premature release was dependent on drug solubility, ionization profile and capsule-fill level. At 100 mg drug-fill level, acetaminophen, trimethoprim and atenolol gave rise to 10.6, 12.2 and 83.1% drug release, respectively, in normal, fasted, gastric fluids. Diffusion layer pH of trimethoprim and atenolol in pH 2 media was determined to be pH 6.3 and 10.3, respectively. Upon increasing capsule-fill load using microcrystalline cellulose as a diluent, a significant reduction in premature gastric release was observed. However, including mannitol as a diluent was only effective at decreasing premature drug release at a low drug-to-diluent ratio. Systematic in vitro screening of enteric capsule fills needs to be conducted to ensure that drug product performance is not compromised.
Collapse
|
15
|
Kalsoom S, Zamir A, Rehman AU, Ashraf W, Imran I, Saeed H, Majeed A, Alqahtani F, Rasool MF. Clinical pharmacokinetics of nadolol: A systematic review. J Clin Pharm Ther 2022; 47:1506-1516. [PMID: 36040016 DOI: 10.1111/jcpt.13764] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/02/2022] [Accepted: 08/10/2022] [Indexed: 11/26/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Nadolol is a non-selective beta-adrenergic antagonist that is used for the treatment of hypertension and angina. The primary route for its administration is oral. It is given once daily as it has a longer half-life (t½). The purpose of conducting this systematic review is to provide a comprehensive view of all the available pharmacokinetic (PK) data on nadolol in humans. This review aimed to systematically collate and analyze publish data on the clinical PK of nadolol in humans and this can be beneficial for the clinicians in dosage adjustments. METHODS Two electronic databases PubMed and Google Scholar were used for conducting a systematic literature search. All the relevant articles containing PK data of nadolol in humans were retrieved. A total of 1275 articles were searched from both databases and after applying eligibility criteria finally, 22 articles were included for conducting the systematic review. RESULTS AND DISCUSSION The area under the plasma concentration curve (AUC) and maximum plasma concentration (Cmax ) of nadolol increased in a dose-dependent manner. The t½ of nadolol was increased to double (18.2-68.6 h) in the patients with chronic kidney disease while the serum t½ became shorter (3.2-4.3 h) when administered to the children. The bioavailability of nadolol was greatly reduced by the coadministration of green tea. Nadolol can be effectively removed by hemodialysis. It undergoes enterohepatic circulation thus activated charcoal decreased its bioavailability. WHAT IS NEW AND CONCLUSION Since, there is no previous report of a systematic review on the PK of nadolol, the current review encompasses all the relevant published articles on nadolol in humans. The analysis and understanding of PK parameters (AUC, Cmax , and t½) of nadolol may be helpful in the development and evaluation of PK models.
Collapse
Affiliation(s)
- Samia Kalsoom
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Ammara Zamir
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Anees Ur Rehman
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Waseem Ashraf
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Imran Imran
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Hamid Saeed
- University College of Pharmacy, Allama Iqbal Campus, University of the Punjab, Lahore, Pakistan
| | - Abdul Majeed
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Muhammad Fawad Rasool
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| |
Collapse
|
16
|
Elsayed MMA, Aboelez MO, Mohamed MS, Mahmoud RA, El-Shenawy AA, Mahmoud EA, Al-Karmalawy AA, Santali EY, Alshehri S, Elsadek MEM, El Hamd MA, Ramadan AEH. Tailoring of Rosuvastatin Calcium and Atenolol Bilayer Tablets for the Management of Hyperlipidemia Associated with Hypertension: A Preclinical Study. Pharmaceutics 2022; 14:pharmaceutics14081629. [PMID: 36015255 PMCID: PMC9412892 DOI: 10.3390/pharmaceutics14081629] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/22/2022] [Accepted: 07/28/2022] [Indexed: 01/20/2023] Open
Abstract
Hyperlipidemia is still the leading cause of heart disease in patients with hypertension. The purpose of this study is to make rosuvastatin calcium (ROS) and atenolol (AT) bilayer tablets to treat coexisting dyslipidemia and hypertension with a single product. ROS was chosen for the immediate-release layer of the constructed tablets, whereas AT was chosen for the sustained-release layer. The solid dispersion of ROS with sorbitol (1:3 w/w) was utilized in the immediate-release layer while hydroxypropyl methylcellulose (HPMC), ethylcellulose (EC), and sodium bicarbonate were incorporated into the floating sustained-release layer. The concentrations of HPMC and EC were optimized by employing 32 full factorial designs to sustain AT release. The bilayer tablets were prepared by the direct compression method. The immediate-release layer revealed that 92.34 ± 2.27% of ROS was released within 60 min at a pH of 1.2. The second sustained-release layer of the bilayer tablets exhibited delayed release of AT (96.65 ± 3.36% within 12 h) under the same conditions. The release of ROS and AT from the prepared tablets was found to obey the non-Fickian diffusion and mixed models (zero-order, Higuchi and Korsmeyer–Peppas), respectively. Preclinical studies using rabbit models investigated the impact of ROS/AT tablets on lipid profiles and blood pressure. A high-fat diet was used to induce obesity in rabbits. Bilayer ROS/AT tablets had a remarkable effect on decreasing the lipid profiles, slowing weight gain, and lowering blood pressure to normal levels when compared to the control group.
Collapse
Affiliation(s)
- Mahmoud M. A. Elsayed
- Department of Pharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, Sohag University, Sohag 82524, Egypt
- Correspondence: (M.M.A.E.); or (M.A.E.H.); Tel.: +20-1227-6604-70 (M.M.A.E.); +966-5541-17991 (M.A.E.H.)
| | - Moustafa O. Aboelez
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sohag University, Sohag 82524, Egypt
| | - Mohamed S. Mohamed
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al Azhar University, Assiut 71524, Egypt
| | - Reda A. Mahmoud
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al Azhar University, Assiut 71524, Egypt
| | - Ahmed A. El-Shenawy
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al Azhar University, Assiut 71524, Egypt
| | - Essam A. Mahmoud
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Ahmed A. Al-Karmalawy
- Department of Pharmaceutical Medicinal Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
| | - Eman Y. Santali
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Sameer Alshehri
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | | | - Mohamed A. El Hamd
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, South Valley University, Qena 83523, Egypt
- Correspondence: (M.M.A.E.); or (M.A.E.H.); Tel.: +20-1227-6604-70 (M.M.A.E.); +966-5541-17991 (M.A.E.H.)
| | - Abd El hakim Ramadan
- Department of Pharmaceutics, Faculty of Pharmacy, Port Said University, Port Said 42515, Egypt
| |
Collapse
|
17
|
Ceschan NE, Scioli-Montoto S, Sbaraglini ML, Ruiz ME, Smyth HD, Bucalá V, Ramírez-Rigo MV. Nebulization of a polyelectrolyte-drug system for systemic hypertension treatment. Eur J Pharm Sci 2022; 170:106108. [DOI: 10.1016/j.ejps.2021.106108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/29/2021] [Accepted: 12/23/2021] [Indexed: 11/03/2022]
|
18
|
Kumar S, Yadav Ravulapalli S, Kumar Tiwari S, Gupta S, Nair AB, Jacob S. Effect of sex and food on the pharmacokinetics of different classes of BCS drugs in rats after cassette administration. Int J Pharm 2021; 610:121221. [PMID: 34695535 DOI: 10.1016/j.ijpharm.2021.121221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/18/2021] [Accepted: 10/18/2021] [Indexed: 11/18/2022]
Abstract
The cassette dosing technique is employed in the drug discovery stage of non-clinical studies to obtain pharmacokinetic data from multiple drug candidates in a single experiment. The objective of the current investigation was to evaluate the effect of sex and food on the selected pharmacokinetic parameters of four biopharmaceutical classification system (BCS) drugs (BCS-I: propranolol, BCS-II: diclofenac, BCS-III: atenolol, and BCS-IV: acetazolamide) utilizing cassette dosing in male and female rats under fed and fasting conditions. Different animal groups were dosed intravenous (i.v) and oral at 1 and 10 mg/kg, respectively, in the form of cassette at a dose of 5 mL/kg. Blood samples were analyzed by liquid chromatography-tandem mass spectrometry. Pharmacokinetics parameters were calculated using Phoenix software version 8.1. A significant increase (p < 0.05) of the area under the plasma concentration-time (AUC0-last) was observed for diclofenac and acetazolamide in females over males after i.v dosing. Additionally, acetazolamide showed greater instantaneous concentration at the time of dosing, and clearance in females (p < 0.05) compared to males after i.v administration. After oral dosing, propranolol exhibited significant variations (p < 0.05) in the maximum drug concentration (Cmax), AUC0-last, the volume of distribution (Vd), and bioavailability in females as compared to males under fed state. Diclofenac showed significant changes (p < 0.05) in AUC0-last, and clearance (Cl) in females as compared to males under fasting and fed state. However, acetazolamide exhibited a significant enhancement (p < 0.05) in AUC0-last, Vd, and Cl in fasting females than the males. The data here illustrates that there is an appreciable difference in AUC and Cmax values exist in male and female rats under fed and fasting conditions administered with the cassette dosing of tested BCS class drugs.
Collapse
Affiliation(s)
- Satish Kumar
- M M College of Pharmacy, M. M. (Deemed to be University), Mullana, Ambala, Haryana 133207, India
| | | | - Sudhir Kumar Tiwari
- Aragen Life Sciences Private Limited, Plot No. 28 A, IDA Nacharam, Hyderabad 500076, India
| | - Sumeet Gupta
- M M College of Pharmacy, M. M. (Deemed to be University), Mullana, Ambala, Haryana 133207, India.
| | - Anroop B Nair
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Shery Jacob
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates
| |
Collapse
|
19
|
All layers matter: Innovative three-dimensional epithelium-stroma-endothelium intestinal model for reliable permeability outcomes. J Control Release 2021; 341:414-430. [PMID: 34871636 DOI: 10.1016/j.jconrel.2021.11.048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/19/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022]
Abstract
Drug development is an ever-growing field, increasingly requesting reliable in vitro tools to speed up early screening phases, reducing the need for animal experiments. In oral delivery, understanding the absorption pattern of a new drug in the small intestine is paramount. Classical two-dimensional (2D) in vitro models are generally too simplistic and do not accurately represent native tissues. The main goal of this work was to develop an advanced three-dimensional (3D) in vitro intestinal model to test absorption in a more reliable manner, by better mimicking the native environment. The 3D model is composed of a collagen-based stromal layer with embedded fibroblasts mimicking the intestinal lamina propria and providing support for the epithelium, composed of enterocytes and mucus-secreting cells. An endothelial layer, surrogating the absorptive capillary network, is also present. The cellular crosstalk between the different cells present in the model is unveiled, disclosing key players, namely those involved in the contraction of collagen by fibroblasts. The developed 3D model presents lower levels of P-glycoprotein (P-gp) and Multidrug Resistance Protein 2 (MRP2) efflux transporters, which are normally overexpressed in traditional Caco-2 models, and are paramount in the absorption of many compounds. This, allied with transepithelial electrical resistance (TEER) values closer to physiological ranges, leads to improved and more reliable permeability outcomes, which are observed when comparing our results with in vivo data.
Collapse
|
20
|
Prediction of plasma concentrations using in silico modelling and simulation approach: Case of Acebutolol. ANNALES PHARMACEUTIQUES FRANÇAISES 2021; 79:530-538. [PMID: 33675740 DOI: 10.1016/j.pharma.2021.02.004] [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: 11/11/2020] [Revised: 02/21/2021] [Accepted: 02/24/2021] [Indexed: 11/21/2022]
Abstract
PURPOSE The aim of this study was to predict the plasma concentrations of acebutolol tablets with different dissolution profiles using computer modelling and evaluating whether they are bioequivalent using simulated population studies. METHODS The dissolution behaviour of acebutolol was studied in the USP Apparatus-II using different dissolution media for pH 1.2, 4.5, and 6.8 at 37±0.5°C. The obtained dissolution data, as well as plasma concentration-time data of the reference product from the literature were used as inputs to build pharmacokinetic model of acebutolol within GastroPlus™ software (version 9.7, Simulations Plus Inc., Lancaster, CA, USA) to simulate the in vivo profiles of the drug. RESULTS The dissolution profiles of the reference product Sectral® 400mg tablets and a locally produced generic product were>85% in 15min in three dissolution media. Simulation results demonstrated that the brand and generic products would show the same in vivo performance. Population simulation results of the ln-transformed 90% confidence interval for the ratio of Cmax, AUC0-t and AUC0-inf values for the two products were within the 80-125% interval, showing to be bioequivalent. CONCLUSION Based on the in vitro results combined with in silico simulations using GastroPlus™, a biowaiver for immediate release acebutolol tablets is justified. Furthermore, computer modelling has shown to be a very intersting tool to prove the bioequivalence for these products.
Collapse
|
21
|
Husain T, Shoaib MH, Ahmed FR, Yousuf RI, Farooqi S, Siddiqui F, Imtiaz MS, Maboos M, Jabeen S. Investigating Halloysite Nanotubes as a Potential Platform for Oral Modified Delivery of Different BCS Class Drugs: Characterization, Optimization, and Evaluation of Drug Release Kinetics. Int J Nanomedicine 2021; 16:1725-1741. [PMID: 33688188 PMCID: PMC7935346 DOI: 10.2147/ijn.s299261] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 02/17/2021] [Indexed: 01/28/2023] Open
Abstract
Purpose This study systematically investigated the potential of four model drugs (verapamil HCl, flurbiprofen, atenolol, and furosemide), each belonging to a different class of Biopharmaceutics Classification Systems (BCS) to be developed into oral modified release dosage forms after loading with halloysite nanotubes (HNTs). Methods The drugs were studied for their loading (mass gain %) by varying solvent system, method, pH, and ratios of loading into the nanotubes using D-optimal split-plot design with the help of Design Expert software. Drug-loaded halloysites were characterized by XRD, DTA, FTIR, SEM, and HPLC-UV-based assay procedures. Dissolution studies were also performed in dissolution media with pH 1.2, 4.5, and 6.8. Moreover, the optimized samples were evaluated under stress stability conditions for determining prospects for the development of oral dosage forms. Results As confirmed with the results of XRD and DTA, the drugs were found to be converted into amorphous form after loading with halloysite (HNTs). The drugs were loaded in the range of ~7–9% for the four drugs, with agitation providing satisfactory and equivalent loading as compared to vacuum plus agitation based reported methods. FTIR results revealed either only weak electrostatic (verapamil HCl and flurbiprofen) or no interaction with the surface structure of the HNTs. The dissolution profiling depicted significantly retarded release of drugs with Fickian diffusion from a polydisperse system as a model that suits well for the development of oral dosage forms. HPLC-UV-based assay indicated that except furosemide (BCS class IV), the other three drugs are quite suitable for development for oral dosage forms. Conclusion The four drugs investigated undergo phase transformation with HNTs. While agitation is an optimum method for loading drugs with various physicochemical attributes into HNTs; solvent system, loading ratios and pH play an important role in the loading efficiency respective to the drug properties. The study supports the capability of developing HNT-based modified release oral dosage forms for drugs with high solubility.
Collapse
Affiliation(s)
- Tazeen Husain
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Muhammad Harris Shoaib
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Farrukh Rafiq Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Rabia Ismail Yousuf
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Sadaf Farooqi
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Fahad Siddiqui
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Muhammad Suleman Imtiaz
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Madiha Maboos
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Sabahat Jabeen
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, 75270, Pakistan
| |
Collapse
|
22
|
Shimazaki S, Kuroda J, Shimomura K, Misaka S. Urinary Excretion of Nadolol as a Possible In Vivo Probe for Drug Interactions Involving P-Glycoprotein. J Clin Pharmacol 2021; 61:799-805. [PMID: 33387374 DOI: 10.1002/jcph.1812] [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: 10/18/2020] [Accepted: 12/30/2020] [Indexed: 11/09/2022]
Abstract
Nadolol is a hydrophilic and nonselective β-adrenoceptor blocker with a bioavailability of 30%, relatively longer half-life, negligible metabolism, and predominant renal excretion. Previous studies have reported that nadolol is a substrate of P-glycoprotein, and the coadministration with itraconazole, a typical P-glycoprotein inhibitor, results in elevated plasma concentrations and cumulative urinary excretion of nadolol. In this study, we assessed whether measurements of urinary-excreted nadolol can be an alternative method of plasma pharmacokinetics for P-glycoprotein-mediated drug interactions in humans. We reanalyzed the pooled data set of plasma concentration and urinary excretion of nadolol from our previous clinical studies in a total of 32 healthy Japanese adults. The area under the plasma concentration-time curve from 0 to infinity (AUC0-∞ ) of nadolol in individual subjects was significantly correlated with the maximum plasma concentration (r = 0.80, P < .01) and the cumulative amount excreted into urine (Ae ) at 4 (r = 0.51, P = .01), 8 (r = 0.63, P < .01), 24 (r = 0.75, P < .01), and 48 (r = 0.77, P < .01) hours. Significant correlations were also observed between the AUC and Ae during the same respective periods. In the drug interactions of nadolol with itraconazole, rifampicin, a well-known P-glycoprotein inducer, or grapefruit juice, there were significant correlations between the differences in AUC0-48 and those in Ae, 0-48 from the controls in individual subjects. These results suggest that the measurements of urinary excretion of nadolol can be employed as a sensitive and reliable alternative to plasma pharmacokinetics for the evaluation of P-glycoprotein-mediated drug interactions.
Collapse
Affiliation(s)
- Sho Shimazaki
- Department of Pharmacy, Fukushima Medical University Hospital, Fukushima, Japan
| | - Junko Kuroda
- Department of Pharmacy, Fukushima Medical University Hospital, Fukushima, Japan
| | - Kenju Shimomura
- Department of Bioregulation and Pharmacological Medicine, School of Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Shingen Misaka
- Department of Pharmacy, Fukushima Medical University Hospital, Fukushima, Japan.,Department of Bioregulation and Pharmacological Medicine, School of Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| |
Collapse
|
23
|
Schlauersbach J, Hanio S, Lenz B, Vemulapalli SPB, Griesinger C, Pöppler AC, Harlacher C, Galli B, Meinel L. Leveraging bile solubilization of poorly water-soluble drugs by rational polymer selection. J Control Release 2020; 330:36-48. [PMID: 33333120 DOI: 10.1016/j.jconrel.2020.12.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/03/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022]
Abstract
Poorly water-soluble drugs frequently solubilize into bile colloids and this natural mechanism is key for efficient bioavailability. We tested the impact of pharmaceutical polymers on this solubilization interplay using proton nuclear magnetic resonance spectroscopy, dynamic light scattering, and by assessing the flux across model membranes. Eudragit E, Soluplus, and a therapeutically used model polymer, Colesevelam, impacted the bile-colloidal geometry and molecular interaction. These polymer-induced changes reduced the flux of poorly water-soluble and bile interacting drugs (Perphenazine, Imatinib) but did not impact the flux of bile non-interacting Metoprolol. Non-bile interacting polymers (Kollidon VA 64, HPMC-AS) neither impacted the flux of colloid-interacting nor colloid-non-interacting drugs. These insights into the drug substance/polymer/bile colloid interplay potentially point towards a practical optimization parameter steering formulations to efficient bile-solubilization by rational polymer selection.
Collapse
Affiliation(s)
- Jonas Schlauersbach
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, DE-97074 Wuerzburg, Germany
| | - Simon Hanio
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, DE-97074 Wuerzburg, Germany
| | - Bettina Lenz
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, DE-97074 Wuerzburg, Germany
| | | | - Christian Griesinger
- Max Planck Institute for Biophysical Chemistry, Am Faßberg 11, DE-37077 Goetingen, Germany
| | - Ann-Christin Pöppler
- Institute of Organic Chemistry, University of Wuerzburg, Am Hubland, DE-97074 Wuerzburg, Germany
| | | | - Bruno Galli
- Novartis Pharma AG, Lichtstrasse 35, CH-4056 Basel, Switzerland
| | - Lorenz Meinel
- Institute for Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, DE-97074 Wuerzburg, Germany; Helmholtz Institute for RNA-based Infection Biology (HIRI), DE-97070 Wuerzburg, Germany.
| |
Collapse
|
24
|
Russell C, Hussain M, Huen D, Rahman AS, Mohammed AR. Profiling gene expression dynamics underpinning conventional testing approaches to better inform pre-clinical evaluation of an age appropriate spironolactone formulation. Pharm Dev Technol 2020; 26:101-109. [PMID: 33078682 DOI: 10.1080/10837450.2020.1839496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
There is a need to accelerate paediatric formulation evaluation and enhance quality of early stage data in drug development to alleviate the information pinch point present between formulation development and clinical evaluation. This present work reports application of DNA microarrays as a high throughput screening tool identifying markers for prediction of bioavailability and formulation driven physiological responses. With a focus on enhancing paediatric medicine provision, an oral liquid spironolactone suspension was formulated addressing a paediatric target product profile. Caco-2 cells cultured on transwell inserts were implemented in transport assays in vitro and DNA microarrays were used to examine gene expression modulation. Wistar rats were used to derive in vivo bioavailability data. In vitro, genomic, and in vivo data sets were concurrently evaluated linking drug transport and the genomic fingerprint generated by spironolactone formulation exposure. Significant changes in gene expression are reported as a result of formulation exposure. These include genes coding for ATP-binding cassette (ABC) transporters, solute carrier (SLC) transporters, cytochrome P450 (CYP) enzymes, and carboxylesterase enzymes. Genomic findings better inform pre-clinical understanding of pharmacokinetic and pharmacodynamic responses to spironolactone and its active metabolites than current in vitro drug transport assays alone.
Collapse
Affiliation(s)
- Craig Russell
- Aston Pharmacy School, Aston University, Birmingham, UK
| | | | - David Huen
- School of Biology, Chemistry and Forensic Science, University of Wolverhampton, Wolverhampton, UK
| | - Ayesha S Rahman
- School of Pharmacy, University of Wolverhampton, Wolverhampton, UK
| | | |
Collapse
|
25
|
Dynamic features of cardiac vector as alternative markers of drug-induced spatial dispersion. J Pharmacol Toxicol Methods 2020; 104:106894. [PMID: 32645483 DOI: 10.1016/j.vascn.2020.106894] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 05/24/2020] [Accepted: 06/24/2020] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The abnormal amplification of ventricular repolarization dispersion (VRD) has long been linked to proarrhythmia risk. Recently, the measure of VRD through electrocardiogram intervals has been strongly questioned. The search for an efficient and non-invasive surrogate marker of drug-induced dispersion effects constitute an urgent research challenge. METHODS Herein, drug-induced ventricular dispersion is generated by d-Sotalol supply in an In-vitro rabbit heart model. A cilindrical chamber simulates the thorax and a multi-electrode net is used to obtain spatial electrocardiographic signals. Cardiac vector dynamics is captured by novel velocity cardiomarkers obtained by quaternion methods. Through statistical analysis and machine learning technics, we compute potential dispersion markers that could define proarrhythmic risk. RESULTS The cardiomarkers with the greatest statistical significance, both obtained from the electrical cardiac vector, were: the QTω, which is the difference between first and last maxima of angular velocity and λ21vT, the roundness of linear velocity. When comparing with the performance of the current standards (89%), this pair was able to correctly separate 21 out of 22 experiments achieving a performance of 95%. Moreover, the QTω computes in a much more robust basis the QT interval, the current index for drug regulation. DISCUSSION These velocity markers circumvent the problems of accuratelly finding the fiducial points such as the always tricky T-wave end. Given the high performance they achieved, it is provided a promising outcome for future applications to the detection of anomalous changes of heterogeneity that may be useful for the purposes of torsadogenic toxicity studies.
Collapse
|
26
|
Liu Y, Li X, Zhang Y, Huang J, Wu Y, Wang L. Considerations for application of biopharmaceutics classification system in chicken: Exemplified by seven drugs classification. J Vet Pharmacol Ther 2020; 43:179-188. [PMID: 32039497 DOI: 10.1111/jvp.12842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/31/2019] [Accepted: 01/19/2020] [Indexed: 01/13/2023]
Abstract
Biopharmaceutics Classification System (BCS) has gained broad acceptance in promoting the development of human drugs. To date, the applicability of existing human BCS criteria has not been evaluated in chickens. The objective of this study was to discuss the feasibility of BCS extrapolation between species and establish a preliminary chicken BCS by classifying seven veterinary commonly used drugs including metronidazole, amoxicillin, sulfamethoxazole, sulfadiazine, ciprofloxacin hydrochloride, doxycycline hydrochloride, and trimethoprim. Firstly, we finished the determination of physiological parameters affecting solubility in chickens, including body temperature, gastrointestinal pH, and the fluid volume in the gastrointestinal tract (GI), and the drug is considered highly soluble in chicken BCS when the highest dose strength is soluble in 20.40 ml (fed) or 6.73 ml (fasted) over the pH range of 1-8 at 41°C. Drug solubility classification was based on dose number calculation. Metronidazol and amoxicillin were classed differently under fed and fasted conditions. Secondly, we discussed the effect of ABC transporters (MDCK vs. MDCK-chAbcb1/Abcg2) and pH (5.5 vs. 7.4) on drug permeability and classification. The drug is classified as highly permeable when its permeability is equal to or greater than metoprolol tartrate. Though ABC transporters and pH significantly affected the permeability values of drugs (p < .05), the permeability classification of the drugs has not been changed except for sulfamethoxazole. This work highlights some of the significant challenges that would be encountered in order to develop a chicken BCS, this valuable information could serve as a helpful tool during chicken drugs development and to minimize the potential risks when developing formulations.
Collapse
Affiliation(s)
- Yang Liu
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiangxiu Li
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yujuan Zhang
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jinhu Huang
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yucheng Wu
- Nanjing No. 13 Middle School, Nanjing, China
| | - Liping Wang
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
27
|
Demir Y. The behaviour of some antihypertension drugs on human serum paraoxonase-1: an important protector enzyme against atherosclerosis. J Pharm Pharmacol 2019; 71:1576-1583. [DOI: 10.1111/jphp.13144] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 06/30/2019] [Indexed: 11/27/2022]
Abstract
Abstract
Objectives
Paraoxonase-1 (PON1) enzyme is related to high-density lipoprotein (HDL), which is calcium dependent. It has essential roles such as protecting LDL against oxidation and detoxification of highly toxic substances. It is a significant risk to reduce the levels of this enzyme in patients with diabetes mellitus, cardiovascular diseases, hyperthyroidism and chronic renal failure.
Methods
Here, it was reported that the purification of human serum PON1 using straightforward methods and determination of the interactions between some antihypertension drugs and the enzyme.
Key finding
It was found that these drugs exhibit potential inhibitor properties for human serum PON1 with IC50 values in the range of 131.40–369.40 μm and Ki values in the range of 56.24 ± 6.75–286.74 ± 28.28 μm. These drugs showed different inhibition mechanisms. It was determined that midodrine and nadolol were exhibited competitive inhibition, but atenolol and pindolol were exhibited non-competitive inhibition.
Conclusion
Usage of these drugs would be hazardous in some cases.
Collapse
Affiliation(s)
- Yeliz Demir
- Department of Chemistry, Faculty of Sciences, Ataturk University, Erzurum, Turkey
- Department of Pharmacy Services, Nihat Delibalta Göle Vocational High School, Ardahan University, Ardahan, Turkey
| |
Collapse
|
28
|
Ngeacharernkul P, Stamatis SD, Kirsch LE. Particle Size Distribution Equivalency as Novel Predictors for Bioequivalence. AAPS PharmSciTech 2018; 19:2787-2800. [PMID: 30117041 DOI: 10.1208/s12249-018-1121-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 07/06/2018] [Indexed: 11/30/2022] Open
Abstract
The use of particle size distribution (PSD) similarity metrics and the development and incorporation of drug release predictions based on PSD properties into PBPK models for various drug administration routes may provide a holistic approach for evaluating the effect of PSD differences on in vitro drug release and bioavailability of disperse systems. The objectives of this study were to provide a rational approach for evaluating the utility of in vitro PSD comparators for predicting bioequivalence for subcutaneously administered test and reference drug emulsions. Two types of in vitro comparators for test and reference emulsion products were evaluated: PSD characterization comparators (overlap metrics, median, and span ratios) and release profile comparators (f2 and various fractional time ratios). A subcutaneous-input PBPK disposition model was developed to simulate blood concentration-time profiles of reference and test emulsion products and pharmacokinetic responses (e.g., AUC, Cmax, and Tmax) were used to determine bioequivalence. A pool of 10,440 pairs of test and reference products was simulated using Monte Carlo experiments. The PSD and release profile comparators were correlated to pass/fail bioequivalence metrics using logistical regression. Based on the use of single in vitro comparators, the f2 method was the best predictor of bioequivalence prediction. The use of combinations of f2 and PSD overlap comparators (e.g., OVL or PROB) improved bioequivalence prediction to about 90%. Simulation procedures used in this study demonstrated a process for developing reliable in vitro BE predictors.
Collapse
|
29
|
Maboos M, Yousuf RI, Shoaib MH, Nasiri I, Hussain T, Ahmed HF, Iffat W. Effect of lipid and cellulose based matrix former on the release of highly soluble drug from extruded/spheronized, sintered and compacted pellets. Lipids Health Dis 2018; 17:136. [PMID: 29885655 PMCID: PMC5994249 DOI: 10.1186/s12944-018-0783-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 05/16/2018] [Indexed: 11/19/2022] Open
Abstract
Background The study was to develop an extended release (ER) encapsulated and compacted pellets of Atenolol using hydrophobic (wax based and polymeric based) and high viscosity grade hydrophilic matrix formers to control the release of this highly water soluble drug by extrusion/spheronization (ES). Atenolol is used for cardiovascular diseases and available as an immediate release (IR) tablet dosage form. The lipids, Carnauba wax (CW), Glyceryl monostearate (GMS) and cellulose based i.e. Hydroxypropyl methylcellulose (HPMC) and Ethyl cellulose (EC) were used in preparing Atenolol ER pellets. Thermal sintering and compaction techniques were also applied to control the burst release of Atenolol. Method For this purpose, thirty-six trial formulations (F1-F36) were designed by Response Surface Methodology (RSM), using Design-Expert 10 software, keeping (HPMC K4M, K15 M & K100 M), (EC 7FP, 10FP & 100FP), waxes (GMS, & CW), their combinations, sintering temperature and duration, as input variables. Dissolution studies were performed in pH, 1.2, 4.5 and 6.8 dissolution media. Drug release kinetics using different models such as zero order, first order, Korsmeyer-Peppas, Hixon Crowell, Baker-Lonsdale and Higuchi kinetics were studied with the help of DDsolver, an excel based add-in program. Results The formulations F35 and F36 showed compliance with Korsmeyer-Peppas Super case II transport model (R2 = 0.975–0.971) in dissolution medium pH 4.5. No drug excipient interaction observed by FTIR. Stereomicroscopy showed that sintered combination pellets, (F35), were highly spherical (AR = 1.061, and sphericity = 0.943). The cross-sectional SEM magnification (at 7000X) of F34 and F35 showed dense cross-linking. The results revealed that the optimized formulations were F35 (sintered pellets) and F36 (compacted pellets) effectively controlling the drug release for 12 h. Conclusion Extended-release encapsulated, and compacted pellets were successfully prepared after the combination of lipids CW (10%) and GMS (20%) with EC (10FP 20% & 100FP 20%). Sintering and compaction, in addition, stabilized the system and controlled the initial burst release of the drug. Extended release (ER) Atenolol is an effective alternative of IR tablets in controlling hypertension and treating other cardiovascular diseases.
Collapse
Affiliation(s)
- Madiha Maboos
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, 75270, Pakistan.,Faculty of Pharmacy, Jinnah University for Women, Karachi, 74600, Pakistan
| | - Rabia Ismail Yousuf
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Muhammad Harris Shoaib
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, 75270, Pakistan.
| | - Iqbal Nasiri
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Tazeen Hussain
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Hafiza Fouzia Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, 75270, Pakistan.,Faculty of Pharmacy, Jinnah University for Women, Karachi, 74600, Pakistan
| | - Wajiha Iffat
- Department of Pharmaceutics, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, 75270, Pakistan
| |
Collapse
|
30
|
Flynn TJ, Vohra SN. Simultaneous determination of intestinal permeability and potential drug interactions of complex mixtures using Caco-2 cells and high-resolution mass spectrometry: Studies with Rauwolfia serpentina extract. Chem Biol Interact 2018; 290:37-43. [PMID: 29782822 DOI: 10.1016/j.cbi.2018.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/30/2018] [Accepted: 05/16/2018] [Indexed: 01/07/2023]
Abstract
Caco-2 cells are a commonly used model for estimating the intestinal bioavailability of single chemical entity pharmaceuticals. Caco-2 cells, when induced with calcitriol, also express other biological functions such as phase I (CYP) and phase II (glucuronosyltransferases) drug metabolizing enzymes which are relevant to drug-supplement interactions. Intestinal bioavailability is an important factor in the overall safety assessment of products consumed orally. Foods, including herbal dietary supplements, are complex substances with multiple chemical components. Because of potential interactions between components of complex mixtures, more reliable safety assessments can be obtained by studying the commercial products "as consumed" rather than by testing individual chemical components one at a time. The present study evaluated the apparent intestinal permeability (Papp) of a model herbal extract, Rauwolfia serpentina, using both whole plant extracts and the individual purified Rauwolfia alkaloids. All test compounds, endpoint substrates, and their metabolites were quantified using liquid chromatography and high-resolution mass spectrometry. The Papp values for individual Rauwolfia alkaloids were comparable whether measured individually or as components of the complete extract. Both Rauwolfia extract and all individual Rauwolfia alkaloids except yohimbine inhibited CYP3A4 activity (midazolam 1'-hydroxylation). Both Rauwolfia extract and all individual Rauwolfia alkaloids except corynanthine and reserpic acid significantly increased glucuronosyltransferase activity (glucuronidation of 4-methylumbelliferone). The positive control, ketoconazole, significantly inhibited both CYP3A4 and glucuronosyltransferase activities. These findings suggest that the Caco-2 assay is capable of simultaneously identifying both bioavailability and potentially hazardous intestinal drug-supplement interactions in complex mixtures.
Collapse
Affiliation(s)
- Thomas J Flynn
- Division of Applied Regulatory Toxicology, U.S. Food and Drug Administration, 8301 Muirkirk Road, Laurel, MD, 20708, USA.
| | - Sanah N Vohra
- Division of Applied Regulatory Toxicology, U.S. Food and Drug Administration, 8301 Muirkirk Road, Laurel, MD, 20708, USA.
| |
Collapse
|
31
|
Functional selectivity of GPCR-directed drug action through location bias. Nat Chem Biol 2017; 13:799-806. [PMID: 28553949 PMCID: PMC5733145 DOI: 10.1038/nchembio.2389] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 03/08/2017] [Indexed: 02/07/2023]
Abstract
G protein-coupled receptors (GPCRs) are increasingly recognized to operate from intracellular membranes as well as the plasma membrane. The β2-adrenergic GPCR can activate Gs-linkedcyclic AMP (cAMP) signaling from endosomes. We show here that the homologous human β1-adrenergic receptor initiates an internal Gs-cAMP signal from the Golgi apparatus. By developing a chemical method to acutely squelch G protein coupling at defined membrane locations, we demonstrate that Golgi activation contributes significantly to the overall cellular cAMP response. Golgi signalling utilizes a pre-existing receptor pool rather than receptors delivered from the cell surface, requiring separate access of extracellular ligands. Epinephrine, a hydrophilic endogenous ligand, accesses the Golgi-localized receptor pool by facilitated transport requiring the organic cation transporter 3 (OCT3) whereas drugs can access the Golgi pool by passive diffusion according to hydrophobicity. We demonstrate marked differences among both agonist and antagonist drugs in Golgi-localized receptor access, and show that β-blocker drugs presently used in the clinic differ markedly in ability to antagonize the Golgi signal. We propose ’location bias’ as a new principle for achieving functional selectivity of GPCR-directed drug action.
Collapse
|
32
|
Kono Y, Iwasaki A, Matsuoka K, Fujita T. Effect of Mechanical Agitation on Cationic Liposome Transport across an Unstirred Water Layer in Caco-2 Cells. Biol Pharm Bull 2017; 39:1293-9. [PMID: 27476939 DOI: 10.1248/bpb.b16-00050] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To develop an effective oral delivery system for plasmid DNA (pDNA) using cationic liposomes, it is necessary to clarify the characteristics of uptake and transport of cationic liposome/pDNA complexes into the intestinal epithelium. In particular, evaluation of the involvement of an unstirred water layer (UWL), which is a considerable permeability barrier, in cationic liposome transport is very important. Here, we investigated the effects of a UWL on the transfection efficiency of cationic liposome/pDNA complexes into a Caco-2 cell monolayer. When Caco-2 cells were transfected with cationic liposome/pDNA complexes in shaking cultures to reduce the thickness of the UWL, gene expression was significantly higher in Caco-2 cells compared with static cultures. We also found that this enhancement of gene expression by shaking was not attributable to activation of transcription factors such as activator protein-1 and nuclear factor-kappaB (NF-κB). In addition, the increase in gene expression by mechanical agitation was observed at all charge ratios (1.5, 2.3, 3.1, 4.5) of cationic liposome/pDNA complexes. Transport experiments using Transwells demonstrated that mechanical agitation increased the uptake of cationic liposome/pDNA complexes by Caco-2 cells, whereas transport of the complexes across a Caco-2 cell monolayer did not occurr. Moreover, the augmentation of the gene expression of cationic liposome/pDNA complexes by shaking was observed in Madin-Darby canine kidney cells. These results indicate that a UWL greatly affects the uptake and transfection efficiency of cationic liposome/pDNA complexes into an epithelial monolayer in vitro.
Collapse
Affiliation(s)
- Yusuke Kono
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University
| | | | | | | |
Collapse
|
33
|
Miyake M, Koga T, Kondo S, Yoda N, Emoto C, Mukai T, Toguchi H. Prediction of drug intestinal absorption in human using the Ussing chamber system: A comparison of intestinal tissues from animals and humans. Eur J Pharm Sci 2017; 96:373-380. [DOI: 10.1016/j.ejps.2016.10.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 08/30/2016] [Accepted: 10/04/2016] [Indexed: 11/30/2022]
|
34
|
Zhang P, Zardán Gómez de la Torre T, Welch K, Bergström C, Strømme M. Supersaturation of poorly soluble drugs induced by mesoporous magnesium carbonate. Eur J Pharm Sci 2016; 93:468-74. [DOI: 10.1016/j.ejps.2016.08.059] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 08/25/2016] [Accepted: 08/29/2016] [Indexed: 11/15/2022]
|
35
|
Zhou W, Johnson TN, Xu H, Cheung S, Bui KH, Li J, Al-Huniti N, Zhou D. Predictive Performance of Physiologically Based Pharmacokinetic and Population Pharmacokinetic Modeling of Renally Cleared Drugs in Children. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2016; 5:475-83. [PMID: 27566992 PMCID: PMC5036422 DOI: 10.1002/psp4.12101] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 06/07/2016] [Accepted: 06/29/2016] [Indexed: 11/17/2022]
Abstract
Predictive performance of physiologically based pharmacokinetic (PBPK) and population pharmacokinetic (PopPK) models of drugs predominantly eliminated through kidney in the pediatric population was evaluated. After optimization using adult clinical data, the verified PBPK models can predict 33 of 34 drug clearance within twofold of the observed values in children 1 month and older. More specifically, 10 of 11 of predicted clearance values were within 1.5‐fold of those observed in children between 1 month and 2 years old. The PopPK approach also predicted 19 of 21 drug clearance within twofold of the observed values in children. In summary, our analysis demonstrated both PBPK and PopPK adult models, after verification with additional adult pharmacokinetic (PK) studies and incorporation of known ontogeny of renal filtration, could be applied for dosing regimen recommendation in children 1 month and older for renally eliminated drugs in a first‐in‐pediatric study.
Collapse
Affiliation(s)
- W Zhou
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | - T N Johnson
- Simcyp (A Certara Company), Sheffield, United Kingdom
| | - H Xu
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | - Sya Cheung
- Quantitative Clinical Pharmacology, AstraZeneca, Cambridge, United Kingdom
| | - K H Bui
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | - J Li
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | - N Al-Huniti
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA
| | - D Zhou
- Quantitative Clinical Pharmacology, AstraZeneca, Waltham, Massachusetts, USA.
| |
Collapse
|
36
|
Exploring the Feasibility of Biowaiver Extension of BCS Class III Drugs with Site-Specific Absorption Using Gastrointestinal Simulation Technology. Eur J Drug Metab Pharmacokinet 2016; 42:471-487. [DOI: 10.1007/s13318-016-0361-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
37
|
Villiger A, Stillhart C, Parrott N, Kuentz M. Using Physiologically Based Pharmacokinetic (PBPK) Modelling to Gain Insights into the Effect of Physiological Factors on Oral Absorption in Paediatric Populations. AAPS JOURNAL 2016; 18:933-47. [DOI: 10.1208/s12248-016-9896-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/21/2016] [Indexed: 12/27/2022]
|
38
|
Misaka S, Knop J, Singer K, Hoier E, Keiser M, Müller F, Glaeser H, König J, Fromm MF. The Nonmetabolized β-Blocker Nadolol Is a Substrate of OCT1, OCT2, MATE1, MATE2-K, and P-Glycoprotein, but Not of OATP1B1 and OATP1B3. Mol Pharm 2016; 13:512-9. [PMID: 26702643 DOI: 10.1021/acs.molpharmaceut.5b00733] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nadolol is a nonmetabolized β-adrenoceptor antagonist and is a substrate of OATP1A2, but not of OATP2B1. However, other drug transporters involved in translocation of nadolol have not been characterized in detail. We therefore investigated nadolol as a potential substrate of the hepatic uptake transporters OATP1B1, OATP1B3, and OCT1 and of the renal transporters OCT2, MATE1, and MATE2-K expressed in HEK cells. Moreover, the importance of P-glycoprotein (P-gp) for nadolol transport was studied using double transfected MDCK-OCT1-P-gp cells. Nadolol was not transported by OATP1B1 and OATP1B3. In contrast, a significantly higher nadolol accumulation (at 1 and 10 μM) was found in OCT1, OCT2, MATE1, and MATE2-K cells compared to control cells (P < 0.01). Km values for OCT2-, MATE1-, and MATE2-K-mediated nadolol uptake were 122, 531, and 372 μM, respectively. Cimetidine (100 μM, P < 0.01) and trimethoprim (100 μM, P < 0.001) significantly inhibited OCT1-, OCT2-, MATE1-, and MATE2-K-mediated nadolol transport. The P-gp inhibitor zosuquidar significantly reduced basal to apical nadolol transport in monolayers of MDCK-OCT1-P-gp cells. In summary, nadolol is a substrate of the cation transporters OCT1, OCT2, MATE1, MATE2-K, and of P-gp. These data will aid future in vivo studies on potential transporter-mediated drug-drug or drug-food interactions with involvement of nadolol.
Collapse
Affiliation(s)
- Shingen Misaka
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen, Germany.,Department of Pharmacology, School of Medicine, Fukushima Medical University , Fukushima, Japan
| | - Jana Knop
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen, Germany
| | - Katrin Singer
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen, Germany
| | - Eva Hoier
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen, Germany
| | - Markus Keiser
- Department of Clinical Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine of Greifswald , Greifswald, Germany
| | - Fabian Müller
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen, Germany
| | - Hartmut Glaeser
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen, Germany
| | - Jörg König
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen, Germany
| | - Martin F Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen, Germany
| |
Collapse
|
39
|
Parr A, Hidalgo IJ, Bode C, Brown W, Yazdanian M, Gonzalez MA, Sagawa K, Miller K, Jiang W, Stippler ES. The Effect of Excipients on the Permeability of BCS Class III Compounds and Implications for Biowaivers. Pharm Res 2015; 33:167-76. [PMID: 26286187 PMCID: PMC4689772 DOI: 10.1007/s11095-015-1773-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 08/10/2015] [Indexed: 11/30/2022]
Abstract
Purpose Currently, the FDA allows biowaivers for Class I (high solubility and high permeability) and Class III (high solubility and low permeability) compounds of the Biopharmaceutics Classification System (BCS). Scientific evidence should be provided to support biowaivers for BCS Class I and Class III (high solubility and low permeability) compounds. Methods Data on the effects of excipients on drug permeability are needed to demonstrate that commonly used excipients do not affect the permeability of BCS Class III compounds, which would support the application of biowaivers to Class III compounds. This study was designed to generate such data by assessing the permeability of four BCS Class III compounds and one Class I compound in the presence and absence of five commonly used excipients. Results The permeability of each of the compounds was assessed, at three to five concentrations, with each excipient in two different models: Caco-2 cell monolayers, and in situ rat intestinal perfusion. No substantial increases in the permeability of any of the compounds were observed in the presence of any of the tested excipients in either of the models, with the exception of disruption of Caco-2 cell monolayer integrity by sodium lauryl sulfate at 0.1 mg/ml and higher. Conclusion The results suggest that the absorption of these four BCS Class III compounds would not be greatly affected by the tested excipients. This may have implications in supporting biowaivers for BCS Class III compounds in general.
Collapse
Affiliation(s)
- Alan Parr
- GlaxoSmithKline Inc., Research Triangle Park, North Carolina, 27709, USA
| | | | - Chris Bode
- Absorption Systems LP, Exton, Pennsylvania, 19341-2556, USA.
| | - William Brown
- US Pharmacopeial Convention, Rockville, Maryland, 20852, USA
| | - Mehran Yazdanian
- Teva Branded Pharmaceuticals R&D Inc., West Chester, Pennsylvania, 19380, USA
| | - Mario A Gonzalez
- P'Kinetics International, Inc., Pembroke Pines, Florida, 33027, USA
| | - Kazuko Sagawa
- Pfizer Global Research and Development, Groton, Connecticut, 06340, USA
| | - Kevin Miller
- GlaxoSmithKline Inc., Research Triangle Park, North Carolina, 27709, USA
| | - Wenlei Jiang
- Food and Drug Administration, Office of Generic Drugs, Silver Spring, Maryland, 20841, USA
| | | |
Collapse
|
40
|
Russell C, Begum S, Hussain Y, Hussain M, Huen D, Rahman AS, Perrie Y, Mohammed AR. Paediatric drug development of ramipril: reformulation,in vitroandin vivoevaluation. J Drug Target 2015; 23:854-63. [DOI: 10.3109/1061186x.2015.1036275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
41
|
Zur M, Cohen N, Agbaria R, Dahan A. The biopharmaceutics of successful controlled release drug product: Segmental-dependent permeability of glipizide vs. metoprolol throughout the intestinal tract. Int J Pharm 2015; 489:304-10. [PMID: 25957705 DOI: 10.1016/j.ijpharm.2015.05.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 04/30/2015] [Accepted: 05/03/2015] [Indexed: 12/20/2022]
Abstract
The purpose of this work was to study the challenges and prospects of regional-dependent absorption in a controlled-release scenario, through the oral biopharmaceutics of the sulfonylurea antidiabetic drug glipizide. The BCS solubility class of glipizide was determined, and its physicochemical properties and intestinal permeability were thoroughly investigated, both in-vitro (PAMPA and Caco-2) and in-vivo in rats. Metoprolol was used as the low/high permeability class boundary marker. Glipizide was found to be a low-solubility compound. All intestinal permeability experimental methods revealed similar trend; a mirror image small intestinal permeability with opposite regional/pH-dependency was obtained, a downward trend for glipizide, and an upward trend for metoprolol. Yet the lowest permeability of glipizide (terminal Ileum) was comparable to the lowest permeability of metoprolol (proximal jejunum). At the colon, similar permeability was evident for glipizide and metoprolol, that was higher than metoprolol's jejunal permeability. We present an analysis that identifies metoprolol's jejunal permeability as the low/high permeability class benchmark anywhere throughout the intestinal tract; we show that the permeability of both glipizide and metoprolol matches/exceeds this threshold throughout the entire intestinal tract, accounting for their success as controlled-release dosage form. This represents a key biopharmaceutical characteristic for a successful controlled-release dosage form.
Collapse
Affiliation(s)
- Moran Zur
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Noa Cohen
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Riad Agbaria
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Arik Dahan
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| |
Collapse
|
42
|
Ozawa M, Tsume Y, Zur M, Dahan A, Amidon GL. Intestinal permeability study of minoxidil: assessment of minoxidil as a high permeability reference drug for biopharmaceutics classification. Mol Pharm 2014; 12:204-11. [PMID: 25423288 DOI: 10.1021/mp500553b] [Citation(s) in RCA: 13] [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
The purpose of this study was to evaluate minoxidil as a high permeability reference drug for Biopharmaceutics Classification System (BCS). The permeability of minoxidil was determined in in situ intestinal perfusion studies in rodents and permeability studies across Caco-2 cell monolayers. The permeability of minoxidil was compared with that of metoprolol, an FDA reference drug for BCS classification. In rat perfusion studies, the permeability of minoxidil was somewhat higher than that of metoprolol in the jejunum, while minoxidil showed lower permeability than metoprolol in the ileum. The permeability of minoxidil was independent of intestinal segment, while the permeability of metoprolol was region-dependent. Similarly, in mouse perfusion study, the jejunal permeability of minoxidil was 2.5-fold higher than that of metoprolol. Minoxidil and metoprolol showed similar permeability in Caco-2 study at apical pH of 6.5 and basolateral pH of 7.4. The permeability of minoxidil was independent of pH, while metoprolol showed pH-dependent transport in Caco-2 study. Minoxidil exhibited similar permeability in the absorptive direction (AP-BL) in comparison with secretory direction (BL-AP), while metoprolol had higher efflux ratio (ER > 2) at apical pH of 6.5 and basolateral pH of 7.4. No concentration-dependent transport was observed for either minoxidil or metoprolol transport in Caco-2 study. Verapamil did not alter the transport of either compounds across Caco-2 cell monolayers. The permeability of minoxidil was independent of both pH and intestinal segment in intestinal perfusion studies and Caco-2 studies. Caco-2 studies also showed no involvement of carrier mediated transport in the absorption process of minoxidil. These results suggest that minoxidil may be an acceptable reference drug for BCS high permeability classification. However, minoxidil exhibited higher jejunal permeability than metoprolol and thus to use minoxidil as a reference drug would raise the permeability criteria for BCS high permeability classification.
Collapse
Affiliation(s)
- Makoto Ozawa
- College of Pharmacy, University of Michigan , 428 Church Street, Ann Arbor, Michigan 48109-1065, United States
| | | | | | | | | |
Collapse
|
43
|
Yu G, Zheng QS, Li GF. Similarities and differences in gastrointestinal physiology between neonates and adults: a physiologically based pharmacokinetic modeling perspective. AAPS JOURNAL 2014; 16:1162-6. [PMID: 25182208 DOI: 10.1208/s12248-014-9652-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/28/2014] [Indexed: 11/30/2022]
Abstract
Physiologically based pharmacokinetic (PBPK) modeling holds great promise for anticipating the quantitative changes of pharmacokinetics in pediatric populations relative to adults, which has served as a useful tool in regulatory reviews. Although the availability of specialized software for PBPK modeling has facilitated the widespread applications of this approach in regulatory submissions, challenges in the implementation and interpretation of pediatric PBPK models remain great, for which controversies and knowledge gaps remain regarding neonatal development of the gastrointestinal tract. The commentary highlights the similarities and differences in the gastrointestinal pH and transit time between neonates and adults from a PBPK modeling prospective. Understanding the similarities and differences in these physiological parameters governing oral absorption would promote good practice in the use of pediatric PBPK modeling to assess oral exposure and pharmacokinetics in neonates.
Collapse
Affiliation(s)
- Guo Yu
- Medical Research Center, Subei People's Hospital, Yangzhou University, No. 98 Nantong West Road, Yangzhou, 225001, China
| | | | | |
Collapse
|
44
|
Zhang Q, Zhang Y, Diamond S, Boer J, Harris JJ, Li Y, Rupar M, Behshad E, Gardiner C, Collier P, Liu P, Burn T, Wynn R, Hollis G, Yeleswaram S. The Janus kinase 2 inhibitor fedratinib inhibits thiamine uptake: a putative mechanism for the onset of Wernicke's encephalopathy. Drug Metab Dispos 2014; 42:1656-62. [PMID: 25063672 DOI: 10.1124/dmd.114.058883] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The clinical development of fedratinib, a Janus kinase (JAK2) inhibitor, was terminated after reports of Wernicke's encephalopathy in myelofibrosis patients. Since Wernicke's encephalopathy is induced by thiamine deficiency, investigations were conducted to probe possible mechanisms through which fedratinib may lead to a thiamine-deficient state. In vitro studies indicate that fedratinib potently inhibits the carrier-mediated uptake and transcellular flux of thiamine in Caco-2 cells, suggesting that oral absorption of dietary thiamine is significantly compromised by fedratinib dosing. Transport studies with recombinant human thiamine transporters identified the individual human thiamine transporter (hTHTR2) that is inhibited by fedratinib. Inhibition of thiamine uptake appears unique to fedratinib and is not shared by marketed JAK inhibitors, and this observation is consistent with the known structure-activity relationship for the binding of thiamine to its transporters. The results from these studies provide a molecular basis for the development of Wernicke's encephalopathy upon fedratinib treatment and highlight the need to evaluate interactions of investigational drugs with nutrient transporters in addition to classic xenobiotic transporters.
Collapse
Affiliation(s)
| | - Yan Zhang
- Incyte Corporation, Wilmington, Delaware
| | | | - Jason Boer
- Incyte Corporation, Wilmington, Delaware
| | | | - Yu Li
- Incyte Corporation, Wilmington, Delaware
| | - Mark Rupar
- Incyte Corporation, Wilmington, Delaware
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Is the full potential of the biopharmaceutics classification system reached? Eur J Pharm Sci 2014; 57:224-31. [DOI: 10.1016/j.ejps.2013.09.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 09/05/2013] [Accepted: 09/15/2013] [Indexed: 11/18/2022]
|
46
|
Ferrie AM, Wang C, Deng H, Fang Y. A label-free optical biosensor with microfluidics identifies an intracellular signalling wave mediated through the β(2)-adrenergic receptor. Integr Biol (Camb) 2014; 5:1253-61. [PMID: 23989552 DOI: 10.1039/c3ib40112j] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The canonical model of G protein-coupled receptor (GPCR) signalling states that it is solely initiated at the cell surface. In recent years, a handful of evidence has started emerging from high-resolution molecular assays that the internalized receptors can mediate the third wave of signalling, besides G protein- and β-arrestin-mediated signalling both initiating at the cell surface. However, little is known about the functional consequences of distinct waves of GPCR signalling, in particular, at the whole cell system level. We here report the development of label-free biosensor antagonist reverse assays and their use to differentiate the signalling waves of an endogenous β2-adrenergic receptor (β2-AR) in A431 cells. Results showed that the persistent agonist treatment activated the β2-ARs, leading to a long-term sustained dynamic mass redistribution (DMR) signal, a whole cell phenotypic response. Under the persistent treatment scheme in microplates, a panel of known β-blockers all dose-dependently and completely reversed the DMR signal of epinephrine at a relatively low dose (10 nM), except for sotalol which partially reversed the DMR. Under the perfusion conditions with microfluidics, the subsequent perfusion with sotalol only reversed the DMR induced by epinephrine or isoproterenol at 10 nM, but not at 10 μM. Furthermore, the degree of the DMR reversion by sotalol was found to be in an opposite relation with the duration of the initial agonist treatment. Together, these results suggest that the hydrophilic antagonist sotalol is constrained outside the cells throughout the assays, and the early signalling wave initiated at the cell surface dominates the DMR induced by epinephrine or isoproterenol at relatively low doses, while a secondary and late signalling wave is initiated once the receptors are internalized and contributes partially to the long-term sustainability of the DMR of epinephrine or isoproterenol at high doses.
Collapse
Affiliation(s)
- Ann M Ferrie
- Biochemical Technologies, Science and Technology Division, Corning Incorporated, Corning, NY 14831, USA.
| | | | | | | |
Collapse
|
47
|
Zur M, Gasparini M, Wolk O, Amidon GL, Dahan A. The low/high BCS permeability class boundary: physicochemical comparison of metoprolol and labetalol. Mol Pharm 2014; 11:1707-14. [PMID: 24735251 DOI: 10.1021/mp500152y] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although recognized as overly conservative, metoprolol is currently the common low/high BCS permeability class boundary reference compound, while labetalol was suggested as a potential alternative. The purpose of this study was to identify the various characteristics that the optimal marker should exhibit, and to investigate the suitability of labetalol as the permeability class reference drug. Labetalol's BCS solubility class was determined, and its physicochemical properties and intestinal permeability were thoroughly investigated, both in vitro and in vivo in rats, considering the complexity of the whole of the small intestine. Labetalol was found to be unequivocally a high-solubility compound. In the pH range throughout the small intestine (6.5-7.5), labetalol exhibited pH-dependent permeability, with higher permeability at higher pH values. While in vitro octanol-buffer partitioning (Log D) values of labetalol were significantly higher than those of metoprolol, the opposite was evident in the in vitro PAMPA permeability assay. The results of the in vivo perfusion studies in rats lay between the two contradictory in vitro studies; metoprolol was shown to have moderately higher rat intestinal permeability than labetalol. Theoretical distribution of the ionic species of the drugs was in corroboration with the experimental in vitro and the in vivo data. We propose three characteristics that the optimal permeability class reference drug should exhibit: (1) fraction dose absorbed in the range of 90%; (2) the optimal marker drug should be absorbed largely via passive transcellular permeability, with no/negligible carrier-mediated active intestinal transport (influx or efflux); and (3) the optimal marker drug should preferably be nonionizable. The data presented in this paper demonstrate that neither metoprolol nor labetalol can be regarded as optimal low/high-permeability class boundary standard. While metoprolol is too conservative due to its complete absorption, labetalol has been shown to be a substrate for P-gp-mediated efflux transport, and both drugs exhibit significant segmental-dependent permeability along the gastrointestinal tract. Nevertheless, the use of metoprolol as the marker compound does not carry a risk of bioinequivalence: Peff value similar to or higher than metoprolol safely indicates high-permeability classification. On the other hand, a more careful data analysis is needed if labetalol is used as the reference compound.
Collapse
Affiliation(s)
- Moran Zur
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev , Beer-Sheva 84105, Israel
| | | | | | | | | |
Collapse
|
48
|
Misaka S, Yatabe J, Müller F, Takano K, Kawabe K, Glaeser H, Yatabe MS, Onoue S, Werba JP, Watanabe H, Yamada S, Fromm MF, Kimura J. Green tea ingestion greatly reduces plasma concentrations of nadolol in healthy subjects. Clin Pharmacol Ther 2014; 95:432-8. [PMID: 24419562 DOI: 10.1038/clpt.2013.241] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 12/11/2013] [Indexed: 11/09/2022]
Abstract
This study aimed to evaluate the effects of green tea on the pharmacokinetics and pharmacodynamics of the β-blocker nadolol. Ten healthy volunteers received a single oral dose of 30 mg nadolol with green tea or water after repeated consumption of green tea (700 ml/day) or water for 14 days. Catechin concentrations in green tea and plasma were determined. Green tea markedly decreased the maximum plasma concentration (C(max)) and area under the plasma concentration-time curve (AUC(0-48)) of nadolol by 85.3% and 85.0%, respectively (P < 0.01), without altering renal clearance of nadolol. The effects of nadolol on systolic blood pressure were significantly reduced by green tea. [(3)H]-Nadolol uptake assays in human embryonic kidney 293 cells stably expressing the organic anion-transporting polypeptides OATP1A2 and OATP2B1 revealed that nadolol is a substrate of OATP1A2 (Michaelis constant (K(m)) = 84.3 μmol/l) but not of OATP2B1. Moreover, green tea significantly inhibited OATP1A2-mediated nadolol uptake (half-maximal inhibitory concentration, IC(50) = 1.36%). These results suggest that green tea reduces plasma concentrations of nadolol possibly in part by inhibition of OATP1A2-mediated uptake of nadolol in the intestine.
Collapse
Affiliation(s)
- S Misaka
- 1] Department of Pharmacology, School of Medicine, Fukushima Medical University, Fukushima, Japan [2] Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - J Yatabe
- Department of Pharmacology, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - F Müller
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - K Takano
- Department of Pharmacology, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - K Kawabe
- Department of Pharmacokinetics and Pharmacodynamics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - H Glaeser
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - M S Yatabe
- Department of Pharmacology, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - S Onoue
- Department of Pharmacokinetics and Pharmacodynamics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - J P Werba
- Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - H Watanabe
- Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - S Yamada
- Department of Pharmacokinetics and Pharmacodynamics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - M F Fromm
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - J Kimura
- Department of Pharmacology, School of Medicine, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
49
|
Khalil F, Läer S. Physiologically based pharmacokinetic models in the prediction of oral drug exposure over the entire pediatric age range-sotalol as a model drug. AAPS JOURNAL 2014; 16:226-39. [PMID: 24399240 PMCID: PMC3933580 DOI: 10.1208/s12248-013-9555-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 12/03/2013] [Indexed: 11/30/2022]
Abstract
In recent years, the increased interest in pediatric research has enforced the role of physiologically based pharmacokinetic (PBPK) models in pediatric drug development. However, an existing lack of published examples contributes to some uncertainties about the reliability of their predictions of oral drug exposure. Developing and validating pediatric PBPK models for oral drug application shall enrich our knowledge about their limitations and lead to a better use of the generated data. This study was conducted to investigate how whole-body PBPK models describe the oral pharmacokinetics of sotalol over the entire pediatric age. Two leading software tools for whole-body PBPK modeling: Simcyp® (Simcyp Ltd, Sheffield, UK) and PK-SIM® (Bayer Technology Services GmbH, Leverkusen, Germany), were used. Each PBPK model was first validated in adults before scaling to children. Model input parameters were collected from the literature and clinical data for 80 children were used to compare predicted and observed values. The results obtained by both models were comparable and gave an adequate description of sotalol pharmacokinetics in adults and in almost all pediatric age groups. Only in neonates, the mean ratio(Obs/Pred) for any PK parameter exceeded a twofold error range, 2.56 (95% confidence interval (CI), 2.10–3.49) and 2.15 (95% CI, 1.77–2.99) for area under the plasma concentration-time curve from the first to the last concentration point and maximal concentration (Cmax) using SIMCYP® and 2.37 (95% CI, 1.76–3.25) for time to reach Cmax using PK-SIM®. The two PBPK models evaluated in this study reflected properly the age-related pharmacokinetic changes and predicted adequately the oral sotalol exposure in children of different ages, except in neonates.
Collapse
Affiliation(s)
- Feras Khalil
- Institute of Clinical Pharmacy and Pharmacotherapy, Heinrich-Heine University of Düsseldorf, Universitaetsstrasse1, Building. 26.22. Room 02.21, 40225, Düsseldorf, Germany
| | | |
Collapse
|
50
|
Misaka S, Miyazaki N, Fukushima T, Yamada S, Kimura J. Effects of green tea extract and (-)-epigallocatechin-3-gallate on pharmacokinetics of nadolol in rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2013; 20:1247-1250. [PMID: 23920278 DOI: 10.1016/j.phymed.2013.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 05/16/2013] [Accepted: 07/02/2013] [Indexed: 06/02/2023]
Abstract
Green tea catechins have been shown to affect the activities of drug transporters in vitro, including P-glycoprotein and organic anion transporting polypeptides. However, it remains unclear whether catechins influence the in vivo disposition of substrate drugs for these transporters. In the present study, we investigated effects of green tea extract (GTE) and (-)-epigallocatechin-3-gallate (EGCG) on pharmacokinetics of a non-selective hydrophilic β-blocker nadolol, which is reported to be a substrate for several drug transporters and is not metabolized by cytochrome P450 enzymes. Male Sprague-Dawley rats received GTE (400 mg/kg), EGCG (150 mg/kg) or saline (control) by oral gavage, 30 min before a single intragastric administration of 10 mg/kg nadolol. Plasma and urinary concentrations of nadolol were determined using high performance liquid chromatography. Pharmacokinetic parameters were estimated by a noncompartmental analysis. Pretreatment with GTE resulted in marked reductions in the maximum concentration (Cmax) and area under the time-plasma concentration curve (AUC) of nadolol by 85% and 74%, respectively, as compared with control. In addition, EGCG alone significantly reduced Cmax and AUC of nadolol. Amounts of nadolol excreted into the urine were decreased by pretreatments with GTE and EGCG, while the terminal half-life of nadolol was not different among groups. These results suggest that the coadministration with green tea catechins, particularly EGCG, causes a significant alteration in the pharmacokinetics of nadolol, possibly through the inhibition of its intestinal absorption mediated by uptake transporters.
Collapse
Affiliation(s)
- S Misaka
- Department of Pharmacology, School of Medicine, Fukushima Medical University, Fukushima, Japan.
| | | | | | | | | |
Collapse
|