1
|
Ueda K, Moseson DE, Taylor LS. Amorphous solubility advantage: Theoretical considerations, experimental methods, and contemporary relevance. J Pharm Sci 2025; 114:18-39. [PMID: 39222748 DOI: 10.1016/j.xphs.2024.08.029] [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: 07/12/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Twenty-five years ago, Hancock and Parks asked a provocative question: "what is the true solubility advantage for amorphous pharmaceuticals?" Difficulties in determining the amorphous solubility have since been overcome due to significant advances in theoretical understanding and experimental methods. The amorphous solubility is now understood to be the concentration after the drug undergoes liquid-liquid or liquid-glass phase separation, forming a water-saturated drug-rich phase in metastable equilibrium with an aqueous phase containing molecularly dissolved drug. While crystalline solubility is an essential parameter impacting the absorption of crystalline drug formulations, amorphous solubility is a vital factor for considering absorption from supersaturating formulations. However, the amorphous solubility of drugs is complex, especially in the presence of formulation additives and gastrointestinal components, and concentration-based measurements may not indicate the maximum drug thermodynamic activity. This review discusses the concept of the amorphous solubility advantage, including a historical perspective, theoretical considerations, experimental methods for amorphous solubility measurement, and the contribution of supersaturation and amorphous solubility to drug absorption. Leveraging amorphous solubility and understanding the associated physicochemical principles can lead to more effective development strategies for poorly water-soluble drugs, ultimately benefiting therapeutic outcomes.
Collapse
Affiliation(s)
- Keisuke Ueda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Dana E Moseson
- Worldwide Research and Development, Pfizer, Inc., Groton, CT 06340, United States
| | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
2
|
Orleanska J, Wiecek W, Majzner K. Investigation of etravirine uptake and distribution in single aortic endothelial cells in vitro using Raman imaging. Analyst 2024; 149:4454-4463. [PMID: 39022813 DOI: 10.1039/d4an00314d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Etravirine (ETV) is an antiretroviral agent that belongs to the class of non-nucleoside reverse transcriptase inhibitors. This study explores the uptake and distribution of ETV in human aortic endothelial cells (HAECs) using Raman spectroscopy combined with chemometrics. The distinctive chemical structure of ETV facilitates tracking of its uptake by observing the Raman band at 2225 cm-1 in the Raman-silent region. The perinuclear distribution pattern in HAECs depends on drug concentration and incubation time. The uptake of ETV is observed within 5 minutes at a concentration of 10 μM, as evidenced by Raman images. Lower ETV concentrations, reflective of those found in human plasma, are detectable in HAECs by applying chemometric methods to Raman spectra from the perinuclear region. The ETV accumulation process is crucial in advancing our understanding of the drug's impact on biochemical alterations within endothelial cells. Additionally, ETV emerges as a promising Raman reporter for marking subcellular compartments, leveraging the 2225 cm-1 band in the cellular Raman silent region. This research contributes valuable insights into the behavior of ETV at the subcellular level, shedding light on its potential applications and impact on subcellular dynamics.
Collapse
Affiliation(s)
- Jagoda Orleanska
- Jagiellonian University in Krakow, Faculty of Chemistry, Department of Chemical Physics, Gronostajowa 2 St, 30-387 Krakow, Poland.
- Jagiellonian University in Krakow, Doctoral School of Exact and Natural Sciences, Prof. St. Lojasiewicza St 11, 30-348 Krakow, Poland
- Jagiellonian University in Krakow, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego St 14, 30-348 Krakow, Poland
| | - Wiktoria Wiecek
- Jagiellonian University in Krakow, Faculty of Chemistry, Department of Chemical Physics, Gronostajowa 2 St, 30-387 Krakow, Poland.
| | - Katarzyna Majzner
- Jagiellonian University in Krakow, Faculty of Chemistry, Department of Chemical Physics, Gronostajowa 2 St, 30-387 Krakow, Poland.
| |
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
|
Oktay AN, Polli JE. Efficiency of single pharmaceutical surfactants to mimic intestinal biorelevant media solubilization and dissolution of etravirine: Comparison of intrinsic and film dissolution models. Eur J Pharm Sci 2024; 196:106746. [PMID: 38499112 DOI: 10.1016/j.ejps.2024.106746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/18/2024] [Accepted: 03/08/2024] [Indexed: 03/20/2024]
Abstract
We understand that quality control dissolution media may best anticipate in vivo product performance by mimicking in vivo media, but preferably involve at most a single pharmaceutical surfactant for routine laboratory use. The objective here was to estimate the concentrations of six pharmaceutical surfactants to mimic etravirine solubility and intrinsic dissolution rate, as well as dissolution rate from a film model, in each Fed State Simulated Intestinal Fluid Version 2 (FeSSIF-V2) and Fasted State Simulated Intestinal Fluid Version 2 (FaSSIF-V2). Solubility studies and colloid sizing measurements were conducted. Results indicate that all six surfactants were more efficient than FeSSIF-V2 or FaSSIF-V2 at solubilizing drug, and also exhibited higher micelle diffusivities than FeSSIF-V2 and FaSSIF-V2 mixed-micelles. The rank-order potency (on mM basis) of the six pharmaceutical surfactants to mimic etravirine solubility in each FeSSIF-V2 and FaSSIF-V2 was: polysorbate 80 (PS80) > polysorbate 20 (PS20) > polyoxyethylene(23) lauryl ether (POE23) > POE10 > hexadecyltrimethylammonium bromide (HEX) > sodium lauryl sulfate (SLS). This rank-order potency was almost the same to mimic drug dissolution rate into each FeSSIF-V2 and FaSSIF-V2, except POE10 > POE23. For the most potent surfactant, PS80, 0.461 mM and 0.140 mM PS80 was estimated to mimic etravirine's solubility and dissolution rate into FeSSIF-V2, respectively, using the intrinsic dissolution model. The low PS80 concentration to mimic dissolution rate reflects the relatively high diffusivity of PS80 micelles, compared to FeSSIF-V2 mixed-micelle diffusivity, which was the case for all six pharmaceutical surfactants. Results are also presented in terms of a film dissolution model for surfactant-mediated dissolution, where dissolution enhancement was less than that in the intrinsic dissolution model, and the film model required lower surfactant concentration than in intrinsic dissolution model to mimic FeSSIF-V2-enhanced dissolution. Findings have promised to identify single pharmaceutical surfactant concentrations that mimic key performance attributes of biorelevant media.
Collapse
Affiliation(s)
- Ayse Nur Oktay
- Department of Pharmaceutical Sciences, University of Maryland, 20 Penn Street, Baltimore, MD 21201, USA; Gulhane Faculty of Pharmacy, Department of Pharmaceutical Technology, University of Health Sciences, Ankara, Turkey
| | - James E Polli
- Department of Pharmaceutical Sciences, University of Maryland, 20 Penn Street, Baltimore, MD 21201, USA.
| |
Collapse
|
5
|
Kambayashi A. In Silico Modeling Approaches Coupled with In Vitro Characterization in Predicting In Vivo Performance of Drug Delivery System Formulations. Mol Pharm 2023; 20:4344-4353. [PMID: 37523273 DOI: 10.1021/acs.molpharmaceut.3c00184] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Optimization of the in vivo performance of dosage forms in humans is essential in developing not only conventional formulations but also drug delivery system (DDS) formulations. Although animal experiments are still useful for these formulations, in silico approaches have become increasingly important for DDS formulations with regard to species-specific differences in physiology that can affect the in vivo performance of dosage forms between animals and humans. Furthermore, it is also important to couple in vitro characterizations with in silico models to predict in vivo performance in humans precisely. In this review article, I summarized in vitro-in silico approaches to predicting the in vivo performance of oral DDS formulations (amorphous solid dispersions, lipid-based formulations, nanosized formulations, cyclodextrins-based formulations, sustained release products, enteric coat products, and orally disintegrating tablets) and parenteral DDS formulations (cyclodextrins-based formulations, liposomes, and inhaled formulations).
Collapse
Affiliation(s)
- Atsushi Kambayashi
- Pharmaceutical Research and Technology Laboratories, Astellas Pharma Incorporated, 180 Ozumi, Yaizu, Shizuoka 425-0072, Japan
| |
Collapse
|
6
|
Reppas C, Kuentz M, Bauer-Brandl A, Carlert S, Dallmann A, Dietrich S, Dressman J, Ejskjaer L, Frechen S, Guidetti M, Holm R, Holzem FL, Karlsson Ε, Kostewicz E, Panbachi S, Paulus F, Senniksen MB, Stillhart C, Turner DB, Vertzoni M, Vrenken P, Zöller L, Griffin BT, O'Dwyer PJ. Leveraging the use of in vitro and computational methods to support the development of enabling oral drug products: An InPharma commentary. Eur J Pharm Sci 2023; 188:106505. [PMID: 37343604 DOI: 10.1016/j.ejps.2023.106505] [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: 03/13/2023] [Revised: 06/18/2023] [Accepted: 06/19/2023] [Indexed: 06/23/2023]
Abstract
Due to the strong tendency towards poorly soluble drugs in modern development pipelines, enabling drug formulations such as amorphous solid dispersions, cyclodextrins, co-crystals and lipid-based formulations are frequently applied to solubilize or generate supersaturation in gastrointestinal fluids, thus enhancing oral drug absorption. Although many innovative in vitro and in silico tools have been introduced in recent years to aid development of enabling formulations, significant knowledge gaps still exist with respect to how best to implement them. As a result, the development strategy for enabling formulations varies considerably within the industry and many elements of empiricism remain. The InPharma network aims to advance a mechanistic, animal-free approach to the assessment of drug developability. This commentary focuses current status and next steps that will be taken in InPharma to identify and fully utilize 'best practice' in vitro and in silico tools for use in physiologically based biopharmaceutic models.
Collapse
Affiliation(s)
- Christos Reppas
- Department of Pharmacy, National and Kapodistrian University of Athens, Greece
| | - Martin Kuentz
- School of Life Sciences, University of Applied Sciences and Arts Northwestern Switzerland, Muttenz CH 4132, Switzerland
| | - Annette Bauer-Brandl
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, Odense 5230, Denmark
| | | | - André Dallmann
- Pharmacometrics/Modeling and Simulation, Research and Development, Pharmaceuticals, Bayer AG, Leverkusen, Germany
| | - Shirin Dietrich
- Department of Pharmacy, National and Kapodistrian University of Athens, Greece
| | - Jennifer Dressman
- Fraunhofer Institute of Translational Medicine and Pharmacology, Frankfurt am Main, Germany
| | - Lotte Ejskjaer
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Sebastian Frechen
- Pharmacometrics/Modeling and Simulation, Research and Development, Pharmaceuticals, Bayer AG, Leverkusen, Germany
| | - Matteo Guidetti
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, Odense 5230, Denmark; Solvias AG, Department for Solid-State Development, Römerpark 2, 4303 Kaiseraugst, Switzerland
| | - René Holm
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, Odense 5230, Denmark
| | - Florentin Lukas Holzem
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, Odense 5230, Denmark; Pharmaceutical R&D, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | | | - Edmund Kostewicz
- Fraunhofer Institute of Translational Medicine and Pharmacology, Frankfurt am Main, Germany
| | - Shaida Panbachi
- School of Life Sciences, University of Applied Sciences and Arts Northwestern Switzerland, Muttenz CH 4132, Switzerland
| | - Felix Paulus
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, Odense 5230, Denmark
| | - Malte Bøgh Senniksen
- Fraunhofer Institute of Translational Medicine and Pharmacology, Frankfurt am Main, Germany; Pharmaceutical R&D, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Cordula Stillhart
- Pharmaceutical R&D, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | | | - Maria Vertzoni
- Department of Pharmacy, National and Kapodistrian University of Athens, Greece
| | - Paul Vrenken
- Department of Pharmacy, National and Kapodistrian University of Athens, Greece; Pharmacometrics/Modeling and Simulation, Research and Development, Pharmaceuticals, Bayer AG, Leverkusen, Germany
| | - Laurin Zöller
- AstraZeneca R&D, Gothenburg, Sweden; Fraunhofer Institute of Translational Medicine and Pharmacology, Frankfurt am Main, Germany
| | | | | |
Collapse
|
7
|
Vinarov Z, Butler J, Kesisoglou F, Koziolek M, Augustijns P. Assessment of food effects during clinical development. Int J Pharm 2023; 635:122758. [PMID: 36801481 DOI: 10.1016/j.ijpharm.2023.122758] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 01/27/2023] [Accepted: 02/17/2023] [Indexed: 02/21/2023]
Abstract
Food-drug interactions frequently hamper oral drug development due to various physicochemical, physiological and formulation-dependent mechanisms. This has stimulated the development of a range of promising biopharmaceutical assessment tools which, however, lack standardized settings and protocols. Hence, this manuscript aims to provide an overview of the general approach and the methodology used in food effect assessment and prediction. For in vitro dissolution-based predictions, the expected food effect mechanism should be carefully considered when selecting the level of complexity of the model, together with its drawbacks and advantages. Typically, in vitro dissolution profiles are then incorporated into physiologically based pharmacokinetic models, which can estimate the impact of food-drug interactions on bioavailability within 2-fold prediction error, at least. Positive food effects related to drug solubilization in the GI tract are easier to predict than negative food effects. Preclinical animal models also provide a good level of food effect prediction, with beagle dogs remaining the gold standard. When solubility-related food-drug interactions have large clinical impact, advanced formulation approaches can be used to improve fasted state pharmacokinetics, hence decreasing the fasted/fed difference in oral bioavailability. Finally, the knowledge from all studies should be combined to secure regulatory approval of the labelling instructions.
Collapse
Affiliation(s)
- Zahari Vinarov
- Department of Chemical and Pharmaceutical Engineering, Sofia University, Sofia, Bulgaria; Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - James Butler
- Medicine Development and Supply, GlaxoSmithKline Research and Development, Ware, United Kingdom
| | | | - Mirko Koziolek
- AbbVie Deutschland GmbH & Co. KG, Small Molecule CMC Development, Ludwigshafen, Germany
| | - Patrick Augustijns
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
8
|
P04-11 Integrated in vivo and in silico (PBPK) study for pharmacokinetic profiling of amorphous solid dispersion of riluzole. Toxicol Lett 2022. [DOI: 10.1016/j.toxlet.2022.07.295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
9
|
Nur Oktay A, Polli JE. Comparison of a single pharmaceutical surfactant versus intestinal biorelevant media for etravirine dissolution: Role and impact of micelle diffusivity. Int J Pharm 2022; 624:122015. [PMID: 35839980 DOI: 10.1016/j.ijpharm.2022.122015] [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: 06/02/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 11/19/2022]
Abstract
Etravirine is an antiviral whose oral absorption is limited by low solubility/dissolution. The objective was to predict and compare etravirine's surfactant-mediated dissolution into polyoxyethylene-10 lauryl ether (POE) and FeSSIF-V2, including the contribution of slow micelle diffusivity. Dynamic light scattering (DLS) was used to measure the size and diffusivity values of drug-loaded micelles. In vitro intrinsic dissolution into surfactant media were predicted using a model for surfactant-mediated dissolution. Compared to maleic buffer, POE and FeSSIF-V2 increased etravirine solubility 232-fold and 8.97-fold, respectively. From DLS, micelle diffusivity of drug-loaded POE micelle and FeSSIF-V2 mixed-micelle was 5.15x10-7 cm2/s and 5.76x10-8 cm2/s, respectively. Observed and predicted dissolution enhancement into POE were 50.7 and 31.3, and 1.26 and 1.24 into FeSSIF-V2, respectively. Hence, there was high dissolution enhancement into POE, although the observed enhancement was only 21.9% of the observed solubility enhancement, reflecting the attenuating impact of the large and slowly diffusing drug-loaded POE micelles. Meanwhile, there was minimal dissolution enhancement into FeSSIF-V2, and the observed enhancement was only 14.0% of the observed solubility enhancement, reflecting the even slower diffusing drug-loaded FeSSIF-V2 mixed-micelles compared to drug-loaded POE micelles. Results are considered in light of designing a single pharmaceutical surfactant system for dissolution that mimics a FeSSIF-V2 system.
Collapse
Affiliation(s)
- Ayse Nur Oktay
- University of Maryland, Department of Pharmaceutical Sciences, 20 Penn Street, Baltimore, MD 21201, USA; University of Health Sciences, Gulhane Faculty of Pharmacy, Department of Pharmaceutical Technology, Ankara, Turkey.
| | - James E Polli
- University of Maryland, Department of Pharmaceutical Sciences, 20 Penn Street, Baltimore, MD 21201, USA
| |
Collapse
|
10
|
Liu L, Chen L, Müllers W, Serno P, Qian F. Water-Resistant Drug-Polymer Interaction Contributes to the Formation of Nano-Species during the Dissolution of Felodipine Amorphous Solid Dispersions. Mol Pharm 2022; 19:2888-2899. [PMID: 35759395 DOI: 10.1021/acs.molpharmaceut.2c00250] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Drug-polymer interactions are of great importance in amorphous solid dispersion (ASD) formulation for both dissolution performance and physical stability considerations. In this work, three felodipine ASD systems with drug loading ranging from 5 to 20% were prepared using PVP, PVP-VA, or HPMC-AS as the polymer matrix. The amorphization and homogeneity were confirmed by differential scanning calorimetry and powder X-ray diffraction. The intrinsic dissolution behavior of these ASDs was studied in 0.05 M HCl and phosphate-buffered saline (PBS) (pH 6.5). In 0.05 M HCl, PVP-VA ASDs with low drug loading (<15%) showed rapid dissolution accompanied with nano-species generation, while in the PVP system, rapid dissolution and nano-species generation were observed only when drug loading was less than 10%, and HPMC-AS ASDs always released slowly with no nano-species formation. In PBS, PVP-VA ASDs with drug loading less than 10% showed rapid dissolution accompanied with nano-species generation, while for PVP ASDs, rapid dissolution and nano-species generation were observed only when drug loading was 5%. However, 20% drug loading HPMC-AS ASDs exhibited rapid dissolution of felodipine and nano-species generation. When the drug loading was above the transition point of PVP-VA ASDs and PVP ASDs, the release rate was significantly lowered, and no nano-species was generated. To understand this phenomenon, drug-polymer interactions were studied using the melting point depression method and the Flory-Huggins model fitting. The Flory-Huggins interaction parameters (χ) for felodipine/HPMC-AS, felodipine/PVP, and felodipine/PVP-VA were determined to be 0.62 ± 0.07, -0.55 ± 0.20, and -1.02 ± 0.21, respectively, indicating the existence of the strongest attractive molecular interaction between felodipine and PVP-VA, followed by felodipine/PVP, but not in felodipine/HPMC-AS. Furthermore, dynamic vapor sorption further revealed that the molecular interactions between felodipine and PVP or PVP-VA were resistant to water. We concluded that water-resistant drug-polymer interactions in felodipine/polymer systems were responsible for the formation of nano-species, which further facilitated the rapid initial drug dissolution.
Collapse
Affiliation(s)
- Lei Liu
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 10084, China
| | - Linc Chen
- Chemical and Pharmaceutical Development, Research and Development, Bayer AG, Beijing 100020, China
| | - Wouter Müllers
- Chemical and Pharmaceutical Development, Research and Development, Bayer AG, Berlin 13342, Germany
| | - Peter Serno
- Chemical and Pharmaceutical Development, Research and Development, Bayer AG, Wuppertal 42096, Germany
| | - Feng Qian
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, and Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 10084, China
| |
Collapse
|
11
|
Exploration of modified cyclone separator for the enhanced recovery of inhalable spray dried cubosomal powder intended to be used for lung delivery. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
12
|
Thakore SD, Sirvi A, Joshi VC, Panigrahi SS, Manna A, Singh R, Sangamwar AT, Bansal AK. Biorelevant dissolution testing and physiologically based absorption modeling to predict in vivo performance of supersaturating drug delivery systems. Int J Pharm 2021; 607:120958. [PMID: 34332060 DOI: 10.1016/j.ijpharm.2021.120958] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 07/08/2021] [Accepted: 07/26/2021] [Indexed: 11/29/2022]
Abstract
Supersaturating drug delivery systems (SDDS) enhance the oral absorption of poorly water-soluble drugs by achieving a supersaturated state in the gastrointestinal tract. The maintenance of a supersaturated state is decided by the complex interplay among inherent properties of drug, excipients and physiological conditions of gastrointestinal tract. The biopharmaceutical advantage through SDDS can be mechanistically investigated by coupling biopredictive dissolution testing with physiologically based absorption modeling (PBAM). However, the development of biopredictive dissolution methods possess challenges due to concurrent dissolution, supersaturation, precipitation, and possible redissolution of precipitates during gastrointestinal transit of SDDS. In this comprehensive review, our effort is to critically assess the current state-of-knowledge and provide future directions for PBAM of SDDS. The review outlines various methods used to retrieve physiologically relevant values for input parameters like solubility, dissolution, precipitation, lipid-digestion and permeability of SDDS. SDDS-specific parameterization includes solubility values corresponding to apparent physical form, dissolution in physiologically relevant volumes with biorelevant media, and transfer experiments to incorporate precipitation kinetics. Interestingly, the lack of experimental permeability values and modification of absorption flux through SDDS possess the additional challenge for its PBAM. Supersaturation triggered permeability modifications are reported to fit the observed plasma concentration-time profile. Hence, the experimental insights on good fitting with modified permeability can be potential area of future research for the development of in vitro methods to reliably predict oral absorption of SDDS.
Collapse
Affiliation(s)
- Samarth D Thakore
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Arvind Sirvi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Vikram C Joshi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Sanjali S Panigrahi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Arijita Manna
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Ridhima Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Abhay T Sangamwar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Arvind K Bansal
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India.
| |
Collapse
|
13
|
Simões MF, Pinto RMA, Simões S. Hot-Melt Extrusion: a Roadmap for Product Development. AAPS PharmSciTech 2021; 22:184. [PMID: 34142250 DOI: 10.1208/s12249-021-02017-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 04/14/2021] [Indexed: 01/01/2023] Open
Abstract
Hot-melt extrusion has found extensive application as a feasible pharmaceutical technological option over recent years. HME applications include solubility enhancement, taste masking, and sustained drug release. As bioavailability enhancement is a hot topic of today's science, one of the main applications of HME is centered on amorphous solid dispersions. This review describes the most significant aspects of HME technology and its use to prepare solid dispersions as a drug formulation strategy to enhance the solubility of poorly soluble drugs. It also addresses molecular and thermodynamic features critical for the physicochemical properties of these systems, mainly in what concerns miscibility and physical stability. Moreover, the importance of applying the Quality by Design philosophy in drug development is also discussed, as well as process analytical technologies in pharmaceutical HME monitoring, under the current standards of product development and regulatory guidance. Graphical Abstract.
Collapse
|
14
|
O'Dwyer PJ, Box KJ, Dressman J, Griffin BT, Henze LJ, Litou C, Pentafragka C, Statelova M, Vertzoni M, Reppas C. Oral biopharmaceutics tools: recent progress from partnership through the Pharmaceutical Education and Research with Regulatory Links collaboration. J Pharm Pharmacol 2021; 73:437-446. [PMID: 33793836 DOI: 10.1093/jpp/rgaa055] [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: 08/25/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVES To summarise key contributions of the Pharmaceutical Education and Research with Regulatory Links (PEARRL) project (2016-2020) to the optimisation of existing and the development of new biopharmaceutics tools for evaluating the in vivo performance of oral drug products during the development of new drugs and at the regulatory level. KEY FINDINGS Optimised biopharmaceutics tools: Based on new clinical data, the composition of biorelevant media for simulating the fed state conditions in the stomach was simplified. Strategies on how to incorporate biorelevant in vitro data of bio-enabling drug products into physiologically based pharmacokinetic (PBPK) modelling were proposed. Novel in vitro biopharmaceutics tools: Small-scale two-stage biphasic dissolution and dissolution-permeation setups were developed to facilitate understanding of the supersaturation effects and precipitation risks of orally administered drugs. A porcine fasted state simulated intestinal fluid was developed to improve predictions and interpretation of preclinical results using in vitro dissolution studies. Based on new clinical data, recommendations on the design of in vitro methodologies for evaluating the GI drug transfer process in the fed state were suggested. The optimized design of in vivo studies for investigating food effects: A food effect study protocol in the pig model was established which successfully predicted the food-dependent bioavailability of two model compounds. The effect of simulated infant fed state conditions in healthy adults on the oral absorption of model drugs was evaluated versus the fasted state and the fed state conditions, as defined by regulatory agencies for adults. Using PBPK modelling, the extrapolated fasted and infant fed conditions data appeared to be more useful to describe early drug exposure in infants, while extrapolation of data collected under fed state conditions, as defined by regulators for adults, failed to capture in vivo infant drug absorption. SUMMARY Substantial progress has been made in developing an advanced suite of biopharmaceutics tools for streamlining drug formulation screening and supporting regulatory applications. These advances in biopharmaceutics were achieved through networking opportunities and research collaborations provided under the H2020 funded PEARRL project.
Collapse
Affiliation(s)
- Patrick J O'Dwyer
- School of Pharmacy, University College Cork, Cork, Ireland.,Pion Inc. (UK) Ltd., Forest Row, East Sussex, UK.,Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| | - Karl J Box
- Pion Inc. (UK) Ltd., Forest Row, East Sussex, UK
| | - Jennifer Dressman
- Institute of Translational Medicine and Pharmacology (ITMP), Fraunhofer Gesellschaft, Frankfurt am Main, Germany
| | | | - Laura J Henze
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Chara Litou
- Institute of Translational Medicine and Pharmacology (ITMP), Fraunhofer Gesellschaft, Frankfurt am Main, Germany
| | - Christina Pentafragka
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| | - Marina Statelova
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| | - Maria Vertzoni
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| | - Christos Reppas
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| |
Collapse
|
15
|
Biorelevant Two-Stage In Vitro Testing for rDCS Classification and in PBPK Modeling–Case Example Ritonavir. J Pharm Sci 2020; 109:2512-2526. [DOI: 10.1016/j.xphs.2020.04.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/30/2020] [Accepted: 04/30/2020] [Indexed: 12/12/2022]
|