1
|
Pepin X, Arora S, Borges L, Cano-Vega M, Carducci T, Chatterjee P, Chen G, Cristofoletti R, Dallmann A, Delvadia P, Dressman J, Fotaki N, Gray E, Heimbach T, Holte Ø, Kijima S, Kotzagiorgis E, Lennernäs H, Lindahl A, Loebenberg R, Mackie C, Malamatari M, McAllister M, Mitra A, Moody R, Mudie D, Musuamba Tshinanu F, Polli JE, Rege B, Ren X, Rullo G, Scherholz M, Song I, Stillhart C, Suarez-Sharp S, Tannergren C, Tsakalozou E, Veerasingham S, Wagner C, Seo P. Parameterization of Physiologically Based Biopharmaceutics Models: Workshop Summary Report. Mol Pharm 2024; 21:3697-3731. [PMID: 38946085 PMCID: PMC11304397 DOI: 10.1021/acs.molpharmaceut.4c00526] [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: 05/13/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 07/02/2024]
Abstract
This Article shares the proceedings from the August 29th, 2023 (day 1) workshop "Physiologically Based Biopharmaceutics Modeling (PBBM) Best Practices for Drug Product Quality: Regulatory and Industry Perspectives". The focus of the day was on model parametrization; regulatory authorities from Canada, the USA, Sweden, Belgium, and Norway presented their views on PBBM case studies submitted by industry members of the IQ consortium. The presentations shared key questions raised by regulators during the mock exercise, regarding the PBBM input parameters and their justification. These presentations also shed light on the regulatory assessment processes, content, and format requirements for future PBBM regulatory submissions. In addition, the day 1 breakout presentations and discussions gave the opportunity to share best practices around key questions faced by scientists when parametrizing PBBMs. Key questions included measurement and integration of drug substance solubility for crystalline vs amorphous drugs; impact of excipients on apparent drug solubility/supersaturation; modeling of acid-base reactions at the surface of the dissolving drug; choice of dissolution methods according to the formulation and drug properties with a view to predict the in vivo performance; mechanistic modeling of in vitro product dissolution data to predict in vivo dissolution for various patient populations/species; best practices for characterization of drug precipitation from simple or complex formulations and integration of the data in PBBM; incorporation of drug permeability into PBBM for various routes of uptake and prediction of permeability along the GI tract.
Collapse
Affiliation(s)
- Xavier Pepin
- Regulatory
Affairs, Simulations Plus Inc., 42505 10th Street West, Lancaster, California 93534-7059, United States
| | - Sumit Arora
- Janssen
Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Luiza Borges
- ANVISA, SIA Trecho 5́, Guara, Brasília, Federal District 71205-050, Brazil
| | - Mario Cano-Vega
- Drug
Product Technologies, Amgen Inc., Thousand Oaks, California 91320-1799, United
States
| | - Tessa Carducci
- Analytical
Commercialization Technology, Merck & Co., Inc., 126 E. Lincoln Ave., Rahway, New Jersey 07065, United States
| | - Parnali Chatterjee
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Grace Chen
- Takeda
Development Center Americas Inc., 300 Shire Way, Lexington, Massachusetts 02421, United States
| | - Rodrigo Cristofoletti
- College
of Pharmacy, University of Florida, 6550 Sanger Rd., Orlando, Florida 32827, United States
| | - André Dallmann
- Bayer
HealthCare SAS, 59000 Lille, France, on behalf of Bayer
AG, Pharmacometrics/Modeling and Simulation, Systems Pharmacology
& Medicine, PBPK, Leverkusen, Germany
| | - Poonam Delvadia
- Office
of Translational Science, Office of Clinical Pharmacology (OCP), Center
for Drug Evaluation and Research (CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United States
| | - Jennifer Dressman
- Fraunhofer Institute of Translational Medicine and Pharmacology, Frankfurt am Main 60596, Germany
| | - Nikoletta Fotaki
- University of Bath, Claverton Down, Bath BA2
7AY, United Kingdom
| | - Elizabeth Gray
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Tycho Heimbach
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Øyvind Holte
- Norwegian Medical Products Agency, Oslo 0213, Norway
| | - Shinichi Kijima
- Office
of New Drug V, Pharmaceuticals and Medical
Devices Agency (PMDA), Tokyo 100-0013, Japan
| | - Evangelos Kotzagiorgis
- European Medicines Agency (EMA), Domenico Scarlattilaan 6, Amsterdam 1083 HS, The Netherlands
| | - Hans Lennernäs
- Translational
Drug Discovery and Development, Department of Pharmaceutical Bioscience, Uppsala University, Uppsala 751 05, Sweden
| | | | - Raimar Loebenberg
- Faculty
of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmontonton T6G 2E1, Canada
| | - Claire Mackie
- Janssen
Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Maria Malamatari
- Medicines & Healthcare Products Regulatory Agency, 10 S Colonnade, London SW1W 9SZ, United Kingdom
| | - Mark McAllister
- Global
Biopharmaceutics, Drug Product Design, Pfizer, Sandwich CT13 9NJ, United Kingdom
| | - Amitava Mitra
- Clinical
Pharmacology, Kura Oncology Inc., Boston, Massachusetts 02210, United States
| | - Rebecca Moody
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Deanna Mudie
- Global
Research and Development, Small Molecules, Lonza, 63045 NE Corporate
Pl., Bend, Oregon 97701, United States
| | - Flora Musuamba Tshinanu
- Belgian Federal Agency for Medicines and Health Products, Galileelaan 5/03, Brussel 1210, Belgium
| | - James E. Polli
- School
of Pharmacy, University of Maryland, Baltimore, Maryland 21201, United States
| | - Bhagwant Rege
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Xiaojun Ren
- PK
Sciences/Translational Medicine, BioMedical Research, Novartis, One Health Plaza, East Hanover, New Jersey 07936, United States
| | - Gregory Rullo
- Regulatory
CMC, AstraZeneca, 1 Medimmune Way, Gaithersburg, Maryland 20878, United States
| | - Megerle Scherholz
- Pharmaceutical
Development, Bristol Myers Squibb, Route 206 & Province Line Road, Princeton, New Jersey 08543, United States
| | - Ivy Song
- Takeda
Development Center Americas Inc., 300 Shire Way, Lexington, Massachusetts 02421, United States
| | - Cordula Stillhart
- Pharmaceutical
R&D, F. Hoffmann-La Roche Ltd., Basel 4070, Switzerland
| | - Sandra Suarez-Sharp
- Regulatory
Affairs, Simulations Plus Inc., 42505 10th Street West, Lancaster, California 93534-7059, United States
| | - Christer Tannergren
- Biopharmaceutics
Science, New Modalities & Parenteral Product Development, Pharmaceutical
Technology & Development, Operations, AstraZeneca, Gothenburg 431 50, Sweden
| | - Eleftheria Tsakalozou
- Division
of Quantitative Methods and Modeling, Office of Research and Standards,
Office of Generic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20903-1058, United
States
| | - Shereeni Veerasingham
- Pharmaceutical
Drugs Directorate (PDD), Health Canada, 1600 Scott St., Ottawa K1A 0K9, Canada
| | - Christian Wagner
- Global
Drug Product Development, Global CMC Development, the Healthcare Business of Merck KGaA, Darmstadt D-64293, Germany
| | - Paul Seo
- Office
of Translational Science, Office of Clinical Pharmacology (OCP), Center
for Drug Evaluation and Research (CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United States
| |
Collapse
|
2
|
Felicijan T, Rakoše I, Prislan M, Locatelli I, Bogataj M, Trontelj J. Application of a Novel Dissolution Medium with Lipids for In Vitro Simulation of the Postprandial Gastric Content. Pharmaceutics 2024; 16:1040. [PMID: 39204385 PMCID: PMC11359312 DOI: 10.3390/pharmaceutics16081040] [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: 07/12/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Food can change various physiological parameters along the gastrointestinal tract, potentially impacting postprandial drug absorption. It is thus important to consider different in vivo conditions during in vitro studies. Therefore, a novel dissolution medium simulating variable postprandial pH values and lipid concentrations was developed and used in this study. Additionally, by establishing and validating a suitable analytical method, the effects of these parameters on the dissolution of a model drug, cinnarizine, and on its distribution between the lipid and aqueous phases of the medium were studied. Both parameters, pH value and lipid concentration, were shown to influence cinnarizine behavior in the in vitro dissolution studies. The amount of dissolved drug decreased with increasing pH due to cinnarizine's decreasing solubility. At pH values 5 and 7, the higher concentration of lipids in the medium increased drug dissolution, and most of the dissolved drug was distributed in the lipid phase. In all media with a lower pH of 3, dissolution was fast and complete, with a significant amount of drug distributed in the lipid phase. These results are in accordance with the in vivo observed positive food effect on cinnarizine bioavailability described in the literature. The developed medium, with its ability to easily adjust the pH level and lipid concentration, thus offers a promising tool for assessing the effect of co-ingested food on the dissolution kinetics of poorly soluble drugs.
Collapse
Affiliation(s)
| | | | | | | | - Marija Bogataj
- Department of Biopharmaceutics and Pharmacokinetics, Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia; (T.F.); (I.R.); (M.P.); (I.L.); (J.T.)
| | | |
Collapse
|
3
|
Danielak D, Gajda M, Bołtromiuk T, Sulikowska K, Kubiak B, Romański M. Drug dissolution and transit in a heterogenous gastric chyme after fed administration: Semi-mechanistic modeling and simulations for an immediate-release and orodispersible tablets containing a poorly soluble drug. Eur J Pharm Biopharm 2024; 200:114341. [PMID: 38795785 DOI: 10.1016/j.ejpb.2024.114341] [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: 02/16/2024] [Revised: 05/06/2024] [Accepted: 05/22/2024] [Indexed: 05/28/2024]
Abstract
Mathematical models that treat the fed stomach content as a uniform entity emptied with a constant rate may not suffice to explain pharmacokinetic profiles recorded in clinical trials. In reality, phenomena such as the Magenstrasse or chyme areas of different pH and viscosity, play an important role in the intragastric drug dissolution and its transfer to the intestine. In this study, we investigated the data gathered in the bioequivalence trial between an immediate-release tablet (Reference) and an orally dispersible tablet (Test) with a poorly soluble weak base drug administered with or without water after a high-fat high-calorie breakfast. Maximum concentrations (Cmax) were significantly greater after administering the Reference product than the Test tablets, despite similar in vitro dissolution profiles. To explain this difference, we constructed a novel semi-mechanistic IVIVP model including a heterogeneous gastric chyme. The drug dissolution in vivo was modeled from the in vitro experiments in biorelevant media simulating gastric and intestinal fluids in the fed state (FEDGAS and FeSSIF). The key novelty of the model was separating the stomach contents into two compartments: isolated chyme (the viscous food content) that carries the drug slowly, and aq_chyme open for rapid Magenstrasse-like routes of drug transit. Drug distribution between these two compartments was both formulation- and administration-dependent, and recognized the respective drug fractions from the clinical pharmacokinetic data. The model's assumption about the nonuniform mixing of the API with the chyme, influencing differential drug dissolution and transit kinetics, led to simulating plasma concentration profiles that reflected well the variability observed in the clinical trial. The model indicated that, after administration, the Reference product mixes to a greater extent with aq_chyme, where the released drug dissolves better and transfers faster to the intestine. In conclusion, this novel approach underlines that diverse gastric emptying of different oral dosage forms may significantly impact pharmacokinetics and affect the outcomes of bioequivalence trials.
Collapse
Affiliation(s)
- Dorota Danielak
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Rokietnicka 3 St, 60-806, Poznań, Poland.
| | - Maciej Gajda
- Adamed Pharma S.A., Pieńków, Mariana Adamkiewicza 6A, 05-152 Czosnów, Poland.
| | - Tomasz Bołtromiuk
- Adamed Pharma S.A., Pieńków, Mariana Adamkiewicza 6A, 05-152 Czosnów, Poland.
| | | | - Bartłomiej Kubiak
- Adamed Pharma S.A., Pieńków, Mariana Adamkiewicza 6A, 05-152 Czosnów, Poland.
| | - Michał Romański
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Rokietnicka 3 St, 60-806, Poznań, Poland.
| |
Collapse
|
4
|
Navas-Bachiller M, Persoons T, D'Arcy DM. In vitro and in silico methods to investigate the effect of moderately increasing medium viscosity and density on ibuprofen dissolution rate. Eur J Pharm Biopharm 2023; 193:74-88. [PMID: 37884158 DOI: 10.1016/j.ejpb.2023.10.018] [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: 12/08/2022] [Revised: 09/18/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
Medium viscosity can affect drug dissolution rate, however, it is not usually considered in routine dissolution testing or less complex biorelevant media. The effects of moderately increasing medium viscosity on the in vitro and in silico dissolution of ibuprofen were investigated with two viscosity enhancing agents (VEA) (hydroxypropyl methylcellulose (HPMC) and sucrose), three viscosity levels (range 0.7-5.5 mPa.s), two solubilities and two fluid velocities in the paddle, flow-through and intrinsic dissolution apparatuses. A factorial design analysis highlighted which factors significantly affected key dissolution metrics. Experimental results in the flow-through apparatus (FTA) were compared with in silico dissolution profiles generated by an in-house simulation code (SIMDISSOTM). Increasing viscosity reduced the intrinsic dissolution rate of ibuprofen for both VEAs. The dissolution rate reduction was also observed in the FTA with sucrose, but less so with HPMC, suggesting particle wetting, motion and surface area effects. Particle motion simulations suggested reduced particle lifting times as viscosity increased, indicating an effect of viscosity on particle dispersal. The viscosity- and fluid density-mediated reduction in the dissolution rate observed with sucrose was accurately simulated by SIMDISSOTM, in particular at higher velocities. Velocity had a significant impact on dissolution rates in the paddle apparatus, with a significant viscosity-related reduction in dissolution observed in the low solubility-low velocity scenario. Even small increases in medium viscosity can reduce the dissolution rate of a BCS class II drug, and in silico particle motion and dissolution data can assist interpretation of particulate dissolution behaviour.
Collapse
Affiliation(s)
- Marina Navas-Bachiller
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland; SSPC, The Science Foundation Ireland Research Centre for Pharmaceuticals, Trinity College Dublin, Ireland.
| | - Tim Persoons
- Department of Mechanical, Manufacturing & Biomedical Engineering, Trinity College Dublin, Ireland; SSPC, The Science Foundation Ireland Research Centre for Pharmaceuticals, Trinity College Dublin, Ireland.
| | - Deirdre M D'Arcy
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland; SSPC, The Science Foundation Ireland Research Centre for Pharmaceuticals, Trinity College Dublin, Ireland.
| |
Collapse
|
5
|
Abdel-Haleem LM, Ramadan NK, El-Rahman MKA, Galal MM. A Screen-Printed Potentiometric Sensor for Stability Indicating Assay and Real-Time Monitoring of Trospium Chloride Dissolution Profile in its Pharmaceutical Dosage Form. JOURNAL OF THE ELECTROCHEMICAL SOCIETY 2023; 170:087506. [DOI: 10.1149/1945-7111/ace8c3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
According to FDA guidance, a biowaiver concept declares that dissolution testing could be approved as a replacement strategy for bioequivalence studies and/or in vivo bioavailability. From the analytical chemistry standpoint, the shift from the classically developed offline methods to the highly integrated miniaturized inline analyzers is one of the pioneering ways that would modernize future of in-vitro - in-vivo correlation (IVIVC). The emergence of screen-printed electrodes (SPE) is now making the move from successive sampling steps and off-line measurements to real-time and in-line monitoring. Recently, “SPE” potentiometric sensor was presented as real-time analyzer that can offer similar analytical results as separation-based chromatographic techniques. Thus, the main objective of this paper is to design a real-time SPE for in situ monitoring of the dissolution of trospium chloride (TRO) in neutral media. Validation of the proposed sensor was performed according to the IUPAC commendations. The measurements performed with this sensor showed an accuracy of average recovery 100.50% and standard deviation of less than 1.0%, also the repeatability and intermediate electrode variabilities were less than 1.0 and 1.3%, respectively. The developed sensor was successfully used for direct observation of the dissolution profile without any need for an extraction step or sample preparation.
Collapse
|
6
|
Cyclodextrin Derivatives as Promising Solubilizers to Enhance the Biological Activity of Rosmarinic Acid. Pharmaceutics 2022; 14:pharmaceutics14102098. [PMID: 36297533 PMCID: PMC9611598 DOI: 10.3390/pharmaceutics14102098] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 11/07/2022] Open
Abstract
Rosmarinic acid (RA) is a natural antioxidant with neuroprotective properties; however, its preventive and therapeutic use is limited due to its slight solubility and poor permeability. This study aimed to improve RA physicochemical properties by systems formation with cyclodextrins (CDs): hydroxypropyl-α-CD (HP-α-CD), HP-β-CD, and HP-γ-CD, which were prepared by the solvent evaporation (s.e.) method. The interactions between components were determined by X-ray powder diffraction (XRPD), differential scanning calorimetry (DSC) and Fourier Transform infrared spectroscopy (FTIR). The sites of interaction between RA and CDs were suggested as a result of in silico studies focused on assessing the interaction between molecules. The impact of amorphous systems formation on water solubility, dissolution rate, gastrointestinal (GIT) permeability, and biological activity was studied. RA solubility was increased from 5.869 mg/mL to 113.027 mg/mL, 179.840 mg/mL, and 194.354 mg/mL by systems formation with HP-α-CD, HP-β-CD, and HP-γ-CD, respectively. During apparent solubility studies, the systems provided an acceleration of RA dissolution. Poor RA GIT permeability at pH 4.5 and 5.8, determined by parallel artificial membrane permeability assay (PAMPA system), was increased; RA–HP-γ-CD s.e. indicated the greatest improvement (at pH 4.5 from Papp 6.901 × 10−7 cm/s to 1.085 × 10−6 cm/s and at pH 5.8 from 5.019 × 10−7 cm/s to 9.680 × 10−7 cm/s). Antioxidant activity, which was determined by DPPH, ABTS, CUPRAC, and FRAP methods, was ameliorated by systems; the greatest results were obtained for RA–HP-γ-CD s.e. The inhibition of acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) was increased from 36.876% for AChE and 13.68% for BChE to a maximum inhibition of the enzyme (plateau), and enabled reaching IC50 values for both enzymes by all systems. CDs are efficient excipients for improving RA physicochemical and biological properties. HP-γ-CD was the greatest one with potential for future food or dietary supplement applications.
Collapse
|
7
|
Doumtsi A, Manousi N, Karavasili C, Fatouros DG, Tzanavaras PD, Zacharis CK. A simple and green LC method for the determination of ibuprofen in milk-containing simulated gastrointestinal media for monitoring the dissolution studies of three dimensional-printed formulations. J Sep Sci 2022; 45:3955-3965. [PMID: 36054076 DOI: 10.1002/jssc.202200444] [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: 06/01/2022] [Revised: 08/31/2022] [Accepted: 08/31/2022] [Indexed: 11/07/2022]
Abstract
A fast and green ultra high-performance LC method was developed for the determination of ibuprofen in milk-containing simulated gastrointestinal media to monitor the dissolution of three-dimensional printed formulations. To remove interfering compounds, protein precipitation using methanol as a precipitation reagent was performed. The separation of the target analyte was performed on an C18 column using a mobile phase consisting of 0.05% v/v aqueous phosphoric acid solution: methanol, 25:75% v/v. Method validation was conducted using the total error concept. The β-expectation tolerance intervals did not exceed the acceptance criteria of ± 15%, meaning that 95% of future results will be included in the defined bias limits. The relative bias ranged between ─ 1.1 to + 3.2% for all analytes, while the relative standard deviation values for repeatability and intermediate precision were less than 2.8% and 3.9%, respectively. The achieved limit of detection was 0.01 μg mL-1 and the lower limit of quantitation was established as 2 μg mL-1 . The proposed method was simple, and it required reduced organic solvent consumption following the requirements of Green Analytical Chemistry. The method was successfully employed for the determination of ibuprofen in real biorelevant media obtained from dissolution studies. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Antigoni Doumtsi
- Laboratory of Pharmaceutical Analysis, Department of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Natalia Manousi
- Laboratory of Analytical Chemistry, Department of Chemistry, Faculty of Sciences, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Christina Karavasili
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Dimitrios G Fatouros
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Paraskevas D Tzanavaras
- Laboratory of Analytical Chemistry, Department of Chemistry, Faculty of Sciences, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Constantinos K Zacharis
- Laboratory of Pharmaceutical Analysis, Department of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| |
Collapse
|
8
|
Manousi N, Karavasili C, Fatouros DG, Tzanavaras PD, Zacharis CK. Development and Validation of an HPLC-UV Method for the Dissolution Studies of 3D-Printed Paracetamol Formulations in Milk-Containing Simulated Gastrointestinal Media. Pharmaceuticals (Basel) 2022; 15:755. [PMID: 35745674 PMCID: PMC9230883 DOI: 10.3390/ph15060755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/11/2022] [Accepted: 06/14/2022] [Indexed: 12/10/2022] Open
Abstract
Herein, a simple and rapid HPLC method for the determination of paracetamol milk-containing biorelevant media is proposed. The separation of the analyte from the milk-containing biorelevant media was accomplished isocratically using a mobile phase containing 25 mM phosphate buffer (pH = 3.0) and methanol, 80:20, v/v at a flow rate of 1 mL min-1. Following a protein precipitation-based sample clean-up, a thorough investigation of the effect of the precipitation reagent (methanol, acetonitrile, 10% v/v trifluoroacetic acid solution) on the analyte recovery was performed. The matrix effect was assessed in each biorelevant medium by comparing the slopes of the calibration curves of aqueous and matrix-matched calibration curves. The method was comprehensively validated using the accuracy profiles. The β-expectation tolerance intervals did not exceed the acceptance criteria of ±15%, meaning that 95% of future results will be included in the defined bias limits. The relative bias ranged between -4.5 and +3.9% for all analytes, while the RSD values for repeatability and intermediate precision were less than 2.7% and 3.0%, respectively. The achieved limit of detection (LOD) was 0.02 μg mL-1 and the lower limits of quantitation (LLOQ) were established as 10 μg mL-1, which corresponded to 2% of the highest expected concentration of paracetamol. The proposed scheme was utilized for the determination of paracetamol in dissolution studies of its 3D-printed formulation in milk-containing biorelevant media.
Collapse
Affiliation(s)
- Natalia Manousi
- Laboratory of Analytical Chemistry, School of Chemistry, Faculty of Sciences, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece; (N.M.); (P.D.T.)
| | - Christina Karavasili
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece; (C.K.); (D.G.F.)
| | - Dimitrios G. Fatouros
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece; (C.K.); (D.G.F.)
| | - Paraskevas D. Tzanavaras
- Laboratory of Analytical Chemistry, School of Chemistry, Faculty of Sciences, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece; (N.M.); (P.D.T.)
| | - Constantinos K. Zacharis
- Laboratory of Pharmaceutical Analysis, Department of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| |
Collapse
|
9
|
Lex TR, Rodriguez JD, Zhang L, Jiang W, Gao Z. Development of In Vitro Dissolution Testing Methods to Simulate Fed Conditions for Immediate Release Solid Oral Dosage Forms. AAPS J 2022; 24:40. [PMID: 35277760 DOI: 10.1208/s12248-022-00690-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 02/10/2022] [Indexed: 11/30/2022] Open
Abstract
In vitro dissolution testing is widely used to mimic and predict in vivo performance of oral drug products in the gastrointestinal (GI) tract. This literature review assesses the current in vitro dissolution methodologies being employed to simulate and predict in vivo drug dissolution under fasted and fed conditions, with emphasis on immediate release (IR) solid oral dosage forms. Notable human GI physiological conditions under fasted and fed states have been reviewed and summarized. Literature results showed that dissolution media, mechanical forces, and transit times are key dissolution test parameters for simulating specific postprandial conditions. A number of biorelevant systems, including the fed stomach model (FSM), GastroDuo device, dynamic gastric model (DGM), simulated gastrointestinal tract models (TIM), and the human gastric simulator (HGS), have been developed to mimic the postprandial state of the stomach. While these models have assisted in expanding physiological relevance of in vitro dissolution tests, in general, these models lack the ability to fully replicate physiological conditions/processes. Furthermore, the translatability of in vitro data to an in vivo system remains challenging. Additionally, physiologically based pharmacokinetic (PBPK) modeling has been employed to evaluate the effect of food on drug bioavailability and bioequivalence. Here, we assess the current status of in vitro dissolution methodologies and absorption PBPK modeling approaches to identify knowledge gaps and facilitate further development of in vitro dissolution methods that factor in fasted and fed states. Prediction of in vivo drug performance under fasted and fed conditions via in vitro dissolution testing and modeling may potentially help efforts in harmonizing global regulatory recommendations regarding in vivo fasted and fed bioequivalence studies for solid oral IR products.
Collapse
Affiliation(s)
- Timothy R Lex
- Division of Complex Drug Analysis, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, St. Louis, Missouri, 63110, USA
| | - Jason D Rodriguez
- Division of Complex Drug Analysis, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, St. Louis, Missouri, 63110, USA
| | - Lei Zhang
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Wenlei Jiang
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, 20993, USA.
| | - Zongming Gao
- Division of Complex Drug Analysis, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, St. Louis, Missouri, 63110, USA.
| |
Collapse
|
10
|
Lemos HD, Prado LD, Rocha HVA. Use of biorelevant dissolution media in dissolution tests as a predictive method of oral bioavailability. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e19759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
11
|
Maroni A, Melocchi A, Zema L, Foppoli A, Gazzaniga A. Retentive drug delivery systems based on shape memory materials. J Appl Polym Sci 2020. [DOI: 10.1002/app.48798] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Alessandra Maroni
- Università degli Studi di Milano, Dipartimento di Scienze Farmaceutiche, Sezione di Tecnologia e Legislazione Farmaceutiche "M.E. Sangalli" Via G. Colombo 71, 20133 Milan Italy
| | - Alice Melocchi
- Università degli Studi di Milano, Dipartimento di Scienze Farmaceutiche, Sezione di Tecnologia e Legislazione Farmaceutiche "M.E. Sangalli" Via G. Colombo 71, 20133 Milan Italy
| | - Lucia Zema
- Università degli Studi di Milano, Dipartimento di Scienze Farmaceutiche, Sezione di Tecnologia e Legislazione Farmaceutiche "M.E. Sangalli" Via G. Colombo 71, 20133 Milan Italy
| | - Anastasia Foppoli
- Università degli Studi di Milano, Dipartimento di Scienze Farmaceutiche, Sezione di Tecnologia e Legislazione Farmaceutiche "M.E. Sangalli" Via G. Colombo 71, 20133 Milan Italy
| | - Andrea Gazzaniga
- Università degli Studi di Milano, Dipartimento di Scienze Farmaceutiche, Sezione di Tecnologia e Legislazione Farmaceutiche "M.E. Sangalli" Via G. Colombo 71, 20133 Milan Italy
| |
Collapse
|
12
|
Segregur D, Flanagan T, Mann J, Moir A, Karlsson EM, Hoch M, Carlile D, Sayah-Jeanne S, Dressman J. Impact of Acid-Reducing Agents on Gastrointestinal Physiology and Design of Biorelevant Dissolution Tests to Reflect These Changes. J Pharm Sci 2019; 108:3461-3477. [DOI: 10.1016/j.xphs.2019.06.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/25/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023]
|
13
|
Bannigan P, Stokes K, Kumar A, Madden C, Hudson SP. Investigating the effects of amphipathic gastrointestinal compounds on the solution behaviour of salt and free base forms of clofazimine: An in vitro evaluation. Int J Pharm 2018; 552:180-192. [DOI: 10.1016/j.ijpharm.2018.09.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 02/02/2023]
|
14
|
Tao C, Yu Y, Chen Z, Zhang M, Liu L, Liu Z, Zhang J, Zhang Q, Song H. Effect of mesopores on solidification of sirolimus self-microemulsifying drug delivery system. CHINESE CHEM LETT 2018. [DOI: 10.1016/j.cclet.2018.11.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
15
|
Koziolek M, Kostewicz E, Vertzoni M. Physiological Considerations and In Vitro Strategies for Evaluating the Influence of Food on Drug Release from Extended-Release Formulations. AAPS PharmSciTech 2018; 19:2885-2897. [PMID: 30155808 DOI: 10.1208/s12249-018-1159-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 08/18/2018] [Indexed: 02/08/2023] Open
Abstract
Food effects on oral drug bioavailability are a consequence of the complex interplay between drug, formulation and human gastrointestinal (GI) physiology. Accordingly, the prediction of the direction and the extent of food effects is often difficult. With respect to novel formulations, biorelevant in vitro methods can be extremely powerful tools to simulate the effect of food-induced changes on the physiological GI conditions on drug release and absorption. However, the selection of suitable in vitro methods should be based on a thorough understanding not only of human GI physiology but also of the drug and formulation properties. This review focuses on in vitro methods that can be applied to evaluate the effect of food intake on drug release from extended release (ER) products during preclinical formulation development. With the aid of different examples, it will be demonstrated that the combined and targeted use of various biorelevant in vitro methods can be extremely useful for understanding drug release from ER products in the fed state and to be able to forecast formulation-associated risks such as dose dumping in early stages of formulation development.
Collapse
|
16
|
Identification of beagle food taking patterns and protocol for food effects evaluation on bioavailability. Sci Rep 2018; 8:12765. [PMID: 30143653 PMCID: PMC6109188 DOI: 10.1038/s41598-018-30937-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 07/17/2018] [Indexed: 12/30/2022] Open
Abstract
Food is a known primary role to the exposure of the drugs orally administered. Since each animal may have unique food taking pattern and it is difficult to manipulate the food taking to animals, there lacks rationalized protocol for the food effects in pre-clinic study. The objective of this study was to identify the beagle food taking patterns and demonstrate their effects on bioavailability in valsartan. Herein, four types of food taking patterns of beagle were identified via inter-day and intra-day analysis, and named as Persisting, Pulsing, Postponing, Pushing (“4P Modes”), respectively, which were also validated by principal component analysis (PCA). Interestingly, food intake resulted in a reduced area under the concentration-time curve (AUC0–12h), maximum concentration (Cmax) and absorption rate, whilst the reduction varied in “4P Modes” of food taking. General considerations in the design of experiment for food effect to the bioavailability in beagles have been established as: to recognize the food taking patterns in each animal, to confirm the inter-day stability of the food taking behaviors, to trace the food taking patterns in parallel with plasma sampling. In conclusion, the right animals with proper food taking patterns should be assessed and selected for pre-clinic bioavailability evaluations.
Collapse
|
17
|
Baxevanis F, Kuiper J, Fotaki N. Impact of presence of excipients in drug analysis in fed-state gastric biorelevant media. Eur J Pharm Biopharm 2018; 131:178-188. [PMID: 30110669 DOI: 10.1016/j.ejpb.2018.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 07/25/2018] [Accepted: 08/12/2018] [Indexed: 01/08/2023]
Abstract
In this study, the influence of the presence of excipients in sample preparation and clean-up steps required prior to drug analysis in milk-based media which simulate the in vivo properties of the fed state stomach was investigated. 15 excipients, normally present in solid dosage forms of five APIs tested (atenolol, paracetamol, furosemide, nifedipine and propafenone hydrochloride) were mixed (one at a time) with the active pharmaceutical ingredient of interest either via vortexing, co-grinding or shaking of the physical mixture and dissolved in Fed State Simulated Gastric Fluid (FeSSGF). The objective of the study was the assessment of the extraction efficiency of three protein precipitation protocols (using MeOH, ΑCN and 10% w/v TCA), typically used in drug analysis, in milk-based biorelevant media in the presence of the excipients. The mixing technique, fat content of the medium and excipient and solvent effects were investigated. The efficiency of three different protein precipitation reagents in drug extraction when dissolved as API:excipient mixtures in the fed-state medium was compared against the equivalent drug amount recovered in the absence of the excipient in FeSSGF. Most excipients had a significant negative effect (p < 0.05) on drug recovery in the milk-based medium as indicated by the multiple linear regression (MLR) analysis performed. For magnesium stearate and HPMC, the % recovery values were the lowest in four out of the five drugs studied, with a range of 10-100% depending on the API, mixing technique and protein precipitation protocol selected. The negative excipient-dependent effect was more profound in nifedipine and propafenone hydrochloride, the most lipophilic compounds of the study. Acetonitrile was the most effective extraction reagent for most drugs in the presence of excipients, followed by methanol and 10% w/v trichloroacetic acid. Data analysis also revealed a dependence of the extraction method efficiency on the medium lipid content. Application of the above extraction protocols in commercially available formulations highlighted the need for assessment of the effect of excipients in extraction efficiency, before transferring the method directly to dissolution studies of formulations in milk-based fed gastric media. In conclusion, the presence of excipients and the selection of protein precipitation protocol are parameters which can affect significantly the efficiency of protein precipitation when FeSSGF is used as dissolution medium and need to be taken into consideration when developing a quantitative method based on the above sample clean-up technique.
Collapse
Affiliation(s)
- Fotios Baxevanis
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Jesse Kuiper
- Analytical Sciences, Merck Research Laboratories, West Point, PA, USA
| | - Nikoletta Fotaki
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK.
| |
Collapse
|
18
|
Baxevanis F, Kuiper J, Fotaki N. Strategic drug analysis in fed-state gastric biorelevant media based on drug physicochemical properties. Eur J Pharm Biopharm 2018; 127:326-341. [DOI: 10.1016/j.ejpb.2018.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/06/2018] [Accepted: 03/02/2018] [Indexed: 12/17/2022]
|
19
|
Lipids in the Stomach – Implications for the Evaluation of Food Effects on Oral Drug Absorption. Pharm Res 2018; 35:55. [DOI: 10.1007/s11095-017-2289-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 10/20/2017] [Indexed: 10/18/2022]
|
20
|
Promzeleva M, Chislov M, Volkova T, Proshin A, Kumeev R, Terekhova I. Effects of Biorelevant Media Components on Dissolution Behaviour of 1,2,4-Thiadiazole Derivative Designed for Alzheimer
ʼs Disease Prevention. Chem Biodivers 2018; 15. [DOI: 10.1002/cbdv.201700459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/13/2017] [Indexed: 11/05/2022]
Affiliation(s)
- Maria Promzeleva
- G.A. Krestov Institute of Solution Chemistry; Russian Academy of Sciences; 1 Akademicheskaya str. 153045 Ivanovo Russia
| | - Mikhail Chislov
- G.A. Krestov Institute of Solution Chemistry; Russian Academy of Sciences; 1 Akademicheskaya str. 153045 Ivanovo Russia
| | - Tatyana Volkova
- G.A. Krestov Institute of Solution Chemistry; Russian Academy of Sciences; 1 Akademicheskaya str. 153045 Ivanovo Russia
| | - Alexey Proshin
- Institute of Physiologically Active Compounds; Russian Academy of Sciences; 1 Severniy pr. 142432 Chernogolovka Russia
| | - Roman Kumeev
- G.A. Krestov Institute of Solution Chemistry; Russian Academy of Sciences; 1 Akademicheskaya str. 153045 Ivanovo Russia
| | - Irina Terekhova
- G.A. Krestov Institute of Solution Chemistry; Russian Academy of Sciences; 1 Akademicheskaya str. 153045 Ivanovo Russia
| |
Collapse
|
21
|
Bou-Chacra N, Melo KJC, Morales IAC, Stippler ES, Kesisoglou F, Yazdanian M, Löbenberg R. Evolution of Choice of Solubility and Dissolution Media After Two Decades of Biopharmaceutical Classification System. AAPS JOURNAL 2017; 19:989-1001. [DOI: 10.1208/s12248-017-0085-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/11/2017] [Indexed: 11/30/2022]
|