1
|
Jain KMH, Duggal I, Hou HH, Siegel RA. Artificial gut Simulator. A scheme to predict intestinal and plasma concentration-time profiles of a weakly basic BCS-II drug, dipyridamole. Eur J Pharm Biopharm 2025; 210:114688. [PMID: 40089075 DOI: 10.1016/j.ejpb.2025.114688] [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: 11/07/2024] [Revised: 02/04/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
The objective of this study was to develop a scheme to predict intestinal and plasma concentration-time profiles of the weakly basic BCS-II drug, dipyridamole (DPD), using an Artificial Gut Simulator (AGS) integrated with a compartment-based disposition model. In vivo data for this study was obtained from previously published literature. A 3-compartment disposition model was developed using the plasma concentration-time profile of DPD following an intravenous bolus dose. The AGS, consisting of a donor cell and a hollow fiber-based absorption module, was tuned to absorb DPD saturated solution at a physiological rate constant, 0.0402 min-1, based on the measured Caco-2 cell monolayer permeability coefficient. The dose dumping technique commonly used during dissolution testing can generate excessively high initial supersaturation and precipitation which is not physiologically relevant. In this study, fractions of DPD dose were added incrementally every 15 min to the AGS donor to simulate an overall first-order gastric emptying process. The concentration absorbed by the hollow fiber receiver media was input into the central compartment of the disposition model. The predicted plasma concentration-time profile matched the human in vivo profile of DPD obtained after oral administration of a 50 mg dose. For 30 and 90 mg oral doses, time profiles of concentration and fraction precipitated in the AGS donor agreed well with human duodenal measurements. This study demonstrates the significance of simulating physiological rate of absorption in vitro to accurately predict the bioavailability of a BCS-II compound.
Collapse
Affiliation(s)
| | - Ishaan Duggal
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Hao Helen Hou
- Small Molecule Pharmaceutical Sciences, Genentech Inc., South San Francisco, CA 94080, USA
| | - Ronald A Siegel
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA; Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
2
|
Ramachandran G, Chacko IA, Mishara MG, Khopade AJ, Sabitha M, Sudheesh MS. A review on design rules for formulating amorphous solid dispersions based on drug-polymer interactions in aqueous environment. Int J Pharm 2025; 675:125541. [PMID: 40164414 DOI: 10.1016/j.ijpharm.2025.125541] [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: 12/23/2024] [Revised: 03/09/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Amorphous solid dispersions (ASDs) are multi-component formulations in which a drug is molecularly dispersed in a carrier. ASDs undergo complex dissolution mechanisms to generate and sustain a supersaturated state of poorly soluble drugs. The link between enhanced solubility, supersaturation stability and drug-polymer interaction (DPI) is critical for the rational design of ASDs. The key mechanism responsible for a high bioavailability is the evolution of supersaturation during the dissolution of ASDs which is also the driving force for drug precipitation. A critical determinant of robust supersaturation generation and stability during dissolution is the molecular interaction between the drug and polymer. Characterization of DPI in a solution state is, however, challenging because of the poor hydrodynamic resolution of the techniques, traditionally used in solid-state analysis. Further, the dissolution conditions, such as the choice of buffer, pH and ionic strength may complicate the analyses and predictions. The role of DPI is a poorly understood aspect of ASD dissolution and therefore is an active area of research. DPI is critical for understanding the design rules for formulating an optimal ASD formulation. The review focuses on different aspects of DPI to stabilize the supersaturated state of a drug during the dissolution of ASDs.
Collapse
Affiliation(s)
- Gayathri Ramachandran
- Molecular Pharmaceutics and Biopharmaceutics Research Lab (MPBRL), Dept. of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi 682041, India
| | - Indhu Annie Chacko
- Molecular Pharmaceutics and Biopharmaceutics Research Lab (MPBRL), Dept. of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi 682041, India
| | - M G Mishara
- Molecular Pharmaceutics and Biopharmaceutics Research Lab (MPBRL), Dept. of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi 682041, India
| | - Ajay Jaysingh Khopade
- Department of Formulation R&D Non-Orals, Sun Pharmaceutical Industries Ltd., Vadodara, India
| | - M Sabitha
- Molecular Pharmaceutics and Biopharmaceutics Research Lab (MPBRL), Dept. of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi 682041, India
| | - M S Sudheesh
- Molecular Pharmaceutics and Biopharmaceutics Research Lab (MPBRL), Dept. of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi 682041, India.
| |
Collapse
|
3
|
Nishiyama S, Takemoto Y, Yamanouchi K, Kondo K, Kawatsu S, Maruyama M, Higaki K. Dynamic changes in the distribution equilibrium of drugs in microemulsions associated with drug absorption facilitate the absorption improvement for drugs with low water-solubility by self-microemulsifying drug delivery system (SMEDDS). Int J Pharm 2025; 674:125458. [PMID: 40074161 DOI: 10.1016/j.ijpharm.2025.125458] [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: 12/19/2024] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
Mechanisms for absorption improvement of drugs with low water-solubility by self-microemulsifying drug delivery system (SMEDDS) are still controversial except for solubility improvement. We attempted to clarify the mechanisms by utilizing model drugs classified as biopharmaceutics classification system class II. In the in-vitro transport study for microemulsions (MEs) formed from SMEDDS, the permeation clearance (CLperm,freeSMEDDS) calculated based on free drug concentrations in MEs, was significantly larger than the CLpermsoln for aqueous solution. However, pretreatment of intestinal mucosa with drug-free MEs did not change CLpermsoln so much. The contribution of endocytosis to drug absorption from MEs was negligible. Instead, our novel egg phosphatidylcholine-monolayer-chloroform partition study revealed that drugs were continuously released from ME droplets, and that the distribution equilibrium of drugs in ME dynamically shifted from ME droplets to aqueous phase associated with their partitioning into chloroform phase (i.e. drug absorption). CLperm,freeSMEDDS did not reflect the continuous drug release or the much larger amount of drugs available for absorption than revealed as free concentrations and thereby overestimated the permeation clearance. The absorption improvement by SMEDDS could be attributed to the dynamic changes in the distribution equilibrium of drugs in MEs associated with drug absorption, i.e., the continuous drug release from ME droplets.
Collapse
Affiliation(s)
- Saki Nishiyama
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Yuki Takemoto
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan; Preformulation Group1, Formulation Development Department, Pharmaceutical Technology Division, Chugai Pharmaceutical Co., Ltd., 216 Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Keita Yamanouchi
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Keiji Kondo
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan; Preformulation Research Laboratory, CMC Headquarters, Otsuka Pharmaceutical Co., Ltd. 224-18 Hiraishi Ebisuno, Kawauchi-cho, Tokushima 771-0182, Japan
| | - Sho Kawatsu
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan; Formulation Design, Pharmaceutical Research and Technology Laboratories, Pharmaceutical Technology, Astellas Pharma Inc., 180 Ozumi, Yaizu, Shizuoka 425-0072, Japan
| | - Masato Maruyama
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Kazutaka Higaki
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.
| |
Collapse
|
4
|
Shetty N, Hau J, Tang S, Chiang PC, Liu J, Jia W, Lubach JW, Nagapudi K, Hou HH. Assessing the impacts of drug loading and polymer type on dissolution behavior and diffusive flux of GDC-6893 amorphous solid dispersions. J Pharm Sci 2025; 114:103686. [PMID: 39880163 DOI: 10.1016/j.xphs.2025.01.021] [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: 10/15/2024] [Revised: 01/12/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025]
Abstract
It is desirable but remains challenging to develop high drug load amorphous solid dispersions (ASDs) without compromising their quality attributes and bio-performance. In this work, we investigated the impacts of formulation variables, such as drug loading (DL) and polymer type, on dissolution behavior, diffusive flux, and in vitro drug absorption of ASDs of a high Tg compound, GDC-6893. ASDs with two polymers (HPMCAS and PVPVA) and various DLs (20 - 80%) were produced by spray drying and their drug-polymer miscibility was evaluated using solid-state nuclear magnetic resonance (ssNMR). μFLUX™ apparatus was used to evaluate the dissolution and drug membrane transport of ASDs at target solution concentrations above the amorphous solubility. Polymer release was monitored using a high-performance liquid chromatography (HPLC) equipped with a charged aerosol detector (CAD). Subsequently, bio-accessibility (%BioA) profiles of the ASDs were evaluated using a benchtop Gastro-Intestinal Model with an advanced gastric compartment (Tiny-TIM), capable of simulating the GI transit as well as in vitro drug dissolution and absorption. Good miscibility and physical stability were observed in ASDs with both HPMCAS and PVPVA even at a high DL of 80%. All GDC-6893 ASDs exhibited dissolution profiles surpassing the amorphous solubility of 20 µg/mL, regardless of the DL and the type of polymer used. Glass-liquid phase separation (GLPS) was observed for ASDs, even at the DL of 80%, and all of these systems reached the maximum achievable diffusive flux. Tiny-TIM results showed an improvement in the %BioA of GDC-6893 ASD compared to its crystalline counterpart however the drug loading and polymer type had no significant impacts on %BioA profiles. Insights from this study suggest that although congruent drug and polymer release was not observed for both HMPCAS- and PVPVA-based ASDs at both 20% and 80% DLs, GDC-6893 and the polymer (HPMCAS or PVPVA) dissolved rapidly from high DL ASDs, followed by the occurrence of GLPS, resulting in the formation of nanosized colloidal species. The findings described herein highlight the importance of understanding both drug and polymer dissolution behavior, as well as in vitro drug absorption, which are essential for the rational design of optimal formulations with desired quality and bio-performance.
Collapse
Affiliation(s)
- Nivedita Shetty
- Department of Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jonathan Hau
- Department of Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Shijia Tang
- Department of Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Po-Chang Chiang
- Department of Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jia Liu
- Department of Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Wei Jia
- Department of Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Joseph W Lubach
- Department of Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Karthik Nagapudi
- Department of Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hao Helen Hou
- Department of Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
5
|
Bordos E, Das G, Schroeder SLM, Florence A, Halbert GW, Robertson J. Probing the interplay between drug saturation, processing temperature and microstructure of amorphous solid dispersions with synchrotron X-ray phase-contrast tomography. Int J Pharm 2025; 669:125018. [PMID: 39626848 DOI: 10.1016/j.ijpharm.2024.125018] [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: 09/18/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
The impact of drug saturation and processing regime on the microstructure of amorphous solid dispersions (ASDs) produced by hot-melt extrusion (HME) has been investigated. By exploring various combinations of drug loadings and processing temperatures, a range of drug saturation points were obtained by HME. The process was monitored with an in-line low-frequency Raman probe to construct the respective solubility phase diagram (i.e., solubility of crystalline drug in molten/soften polymer). The resulting ASDs were analysed with synchrotron X-ray phase-contrast micro computed tomography (Sync-XPC-μCT) in conjunction with a tailored image segmentation strategy to extract quantitative and qualitative descriptors. Despite minimal elemental variability between the drug (paracetamol) and the polymer (HPMC), Sync-XPC-μCT provided sufficient contrast to identify multiple structural domains, including drug-rich crystalline clusters, impurities, polymer-related heterogeneities and voids/pores. Supersaturated ASDs (> 20 wt% drug loading) displayed higher structural complexity and showed a plethora of highly defective API-rich crystalline domains upon ageing, which were absent in the undersaturated ASDs. Beyond its impact on the API physical state, the HME processing regime influenced the degree of homogeneity within the polymer fraction, as well as total porosity, size, shape and pore connectivity. By correlating with fundamental API-polymer solubility data, this study offers additional insight into the dynamics of the drug's solubilisation process during extrusion and the subsequent formation of microstructures within the ASD system, which have potential implications on product performance and stability.
Collapse
Affiliation(s)
- Ecaterina Bordos
- CMAC, University of Strathclyde, Technology and Innovation Centre, 99 George Street, Glasgow, G1 1RD, UK; Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| | - Gunjan Das
- School of Chemical and Process Engineering, University of Leeds, Leeds, UK; Diamond Light Source, Harwell Science and Innovation Campus, Chilton, Didcot, OX11 0DE, UK; EPSRC Future Manufacturing Research Hub, CMAC, Research Complex at Harwell, Harwell Science and Innovation Campus, Chilton, Didcot, OX11 0FA, UK
| | - Sven L M Schroeder
- School of Chemical and Process Engineering, University of Leeds, Leeds, UK; Diamond Light Source, Harwell Science and Innovation Campus, Chilton, Didcot, OX11 0DE, UK; EPSRC Future Manufacturing Research Hub, CMAC, Research Complex at Harwell, Harwell Science and Innovation Campus, Chilton, Didcot, OX11 0FA, UK
| | - Alastair Florence
- CMAC, University of Strathclyde, Technology and Innovation Centre, 99 George Street, Glasgow, G1 1RD, UK; Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Gavin W Halbert
- CMAC, University of Strathclyde, Technology and Innovation Centre, 99 George Street, Glasgow, G1 1RD, UK; Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - John Robertson
- CMAC, University of Strathclyde, Technology and Innovation Centre, 99 George Street, Glasgow, G1 1RD, UK; Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| |
Collapse
|
6
|
Patel RP, Taylor LS, Polli JE. Impact of drug incorporation into micelle on reduced griseofulvin and meloxicam permeation across a hollow fiber membrane. J Pharm Sci 2025; 114:402-415. [PMID: 39426562 DOI: 10.1016/j.xphs.2024.10.017] [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: 06/08/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
A hollow fiber membrane (HFM) was previously characterized as a potential permeation component of a dissolution/permeation system. Two objectives were to assess the impact of micellization on drug permeation across HFM and identify a preferred permeation model from three models: permeation from only free drug, permeation from both free drug and micelle-bound drug, and permeation with enhancement from micelle shuttling. HFM studies were conducted under unsaturated drug conditions, using griseofulvin and the more hydrophilic drug meloxicam, with and without surfactant [sodium lauryl sulfate, polysorbate 80, and polyoxyethylene (10) lauryl ether]. Griseofulvin was micelle incorporated to a greater extent than meloxicam, such that griseofulvin flux decreased to a greater extent than for meloxicam. The griseofulvin permeation model from only free drug was rejected, since griseofulvin flux required free drug to be about 5-20 fold higher in HFM flux studies than supported by solubility studies, depending on surfactant. Permeation from both free griseofulvin and micelle-bound griseofulvin successfully accommodated observed flux, where micelle permeability was about 5-fold lower than free drug permeability for HFM with 10 KDa MWCO. Permeation with enhancement from micelle shuttling was not the preferred explanation, although the model accommodated flux data and provided aqueous boundary layer thicknesses similar to other setups.
Collapse
Affiliation(s)
- Roshni P Patel
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - James E Polli
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA.
| |
Collapse
|
7
|
Hate SS, Thompson SA, Singaraju AB. Impact of sink conditions on drug release behavior of controlled-release formulations. J Pharm Sci 2025; 114:520-529. [PMID: 39481474 DOI: 10.1016/j.xphs.2024.10.032] [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: 09/27/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 11/02/2024]
Abstract
Developing a controlled release (CR) formulations is a complex and iterative process, often requiring preclinical or clinical studies to establish in vitro-in vivo correlations. This can be particularly challenging for poorly soluble drugs due to the non-sink conditions encountered in vitro. Although compendial dissolution methods (e.g., USP II, IV) have historically been used to understand the dissolution performance of CR formulations, there is increasing interest in more physiologically relevant experimental techniques to improve the predictive ability. In this study, traditional USP apparatus as well as the biorelevant absorptive dissolution apparatus were employed to understand the impact of apparatus type and sink condition on the release mechanisms of CR formulations and in turn evaluate the application of absorptive dissolution apparatus for dissolution testing of CR formulations. Release mechanisms were further analyzed using the Peppas equations, providing additional mechanistic insights. The release behavior showed a strong dependence on sink conditions for drugs with low intrinsic solubility, while highly soluble drugs were unaffected by dissolution conditions. Interestingly, the dissolution mechanism was found to be independent of the apparatus type. The study clearly underscores the importance of considering the sink conditions in developing more predictive and biorelevant dissolution testing methods for CR formulations. Furthermore, the study highlights the potential impact on the sink and resultant differences in the drug release mechanisms as a function of the dose.
Collapse
Affiliation(s)
- Siddhi S Hate
- Synthetic Molecule Design and Development, Eli Lilly and Company, Indianapolis, IN 46285, USA.
| | - Stephen A Thompson
- Division of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, TX 78712, USA
| | - Aditya B Singaraju
- Synthetic Molecule Design and Development, Eli Lilly and Company, Indianapolis, IN 46285, USA
| |
Collapse
|
8
|
Elkhabaz A, Moseson DE, Brouwers J, Augustijns P, Taylor LS. Dissolution, phase behavior and mass transport of amorphous solid dispersions in aspirated human intestinal fluids. J Pharm Sci 2025; 114:336-349. [PMID: 39419479 DOI: 10.1016/j.xphs.2024.10.005] [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: 08/09/2024] [Revised: 10/05/2024] [Accepted: 10/06/2024] [Indexed: 10/19/2024]
Abstract
Amorphous solid dispersions (ASDs) typically show improved dissolution and generate supersaturated solutions, enhancing the oral bioavailability of poorly soluble drugs. To gain insights into intraluminal ASD behavior, we utilized two poorly soluble drugs with different crystallization tendencies, atazanavir and posaconazole, prepared as ASDs at a 10% drug loading with hydroxypropyl methylcellulose acetyl succinate (HPMCAS). We evaluated their release in aspirated fasted-state human intestinal fluid (FaHIF), and multi-component fasted-state simulated intestinal fluid (composite-FaSSIF), characterizing the supersaturation profiles and drug-rich nanodroplets that formed. Complete release was observed for atazanavir ASDs over a 90 min period. Flux for dissolved atazanavir ASDs remained high over the experimental time period of 3 h. In contrast, posaconazole solution concentrations were initially high and then decreased. Likewise, flux was initially high and then decreased where these changes are attributed to crystallization of the drug. Generation of spherical nano-sized amorphous droplets of ∼100-150 nm was found to occur in ex vivo FaHIF media for both ASDs, maximizing the diffusive flux during the supersaturation window. Moreover, buffer capacity differences were postulated to influence release rates of ASDs in simulated vs aspirated fluids. Importantly, the solution phase phenomena observed during ASD release in simulated fluids, namely amorphous nanodroplet formation and drug crystallization, were also found to occur in aspirated luminal fluids.
Collapse
Affiliation(s)
- Ahmed Elkhabaz
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
| | - Dana E Moseson
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA
| | | | | | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana, USA.
| |
Collapse
|
9
|
Moseson DE, Li N, Rantanen J, Ueda K, Zhang GGZ. Professor Lynne S. Taylor: Scientist, educator, and adventurer. J Pharm Sci 2025; 114:2-9. [PMID: 39426563 DOI: 10.1016/j.xphs.2024.10.015] [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: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
This special edition of the Journal of Pharmaceutical Sciences is dedicated to Professor Lynne S. Taylor (Retter Distinguished Professor of Pharmacy, Department of Industrial and Molecular Pharmaceutics, Purdue University), to honor her distinguished career as a pharmaceutical scientist and educator. The goal of this commentary is to provide an overview of Professor Taylor's career path, summarize her key research contributions, and provide some insight into her personal and professional contributions as an educator, mentor, wife, mother, friend, and adventurer.
Collapse
Affiliation(s)
- Dana E Moseson
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States.
| | - Na Li
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Jukka Rantanen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Keisuke Ueda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Geoff G Z Zhang
- ProPhysPharm LLC, Lincolnshire, Illinois 60069, United States; Department of Industrial and Molecular Pharmaceutics, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
10
|
B S, Ghosh A. Mechanistic Insights into Amorphous Solid Dispersions: Bridging Theory and Practice in Drug Delivery. Pharm Res 2025; 42:1-23. [PMID: 39849216 DOI: 10.1007/s11095-024-03808-w] [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: 11/01/2024] [Accepted: 12/12/2024] [Indexed: 01/25/2025]
Abstract
Improving the bioavailability of poorly water-soluble drugs presents a significant challenge in pharmaceutical development. Amorphous solid dispersions (ASDs) have garnered substantial attention for their capability to augment the solubility and dissolution rate of poorly water-soluble drugs, thereby markedly enhancing their bioavailability. ASDs, characterized by a metastable equilibrium where the active pharmaceutical ingredient (API) is molecularly dispersed, offer enhanced absorption compared to crystalline forms. This review explores recent research advancements in ASD, emphasizing dissolution mechanisms, phase separation phenomena, and the importance of drug loading and congruency limits on ASD performance. Principal occurrences such as liquid-liquid phase separation (LLPS) and supersaturation are discussed, highlighting their impact on drug solubility, absorption and subsequent bioavailability. Additionally, it addresses the role of polymers in controlling supersaturation, stabilizing drug-rich nanodroplets, and inhibiting recrystallization. Recent advancements and emerging technologies offer new avenues for ASD characterization and production and demonstrate the potential of ASDs to enhance bioavailability and reduce variability, making possible for more effective and patient-friendly pharmaceutical formulations. Future research directions are proposed, focusing on advanced computational models for predicting ASD stability, use of novel polymeric carriers, and methods for successful preparations.
Collapse
Affiliation(s)
- Srividya B
- Solid State Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India
| | - Animesh Ghosh
- Solid State Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India.
| |
Collapse
|
11
|
Qian K, Stella L, Liu F, Jones DS, Andrews GP, Tian Y. Kinetic and Thermodynamic Interplay of Polymer-Mediated Liquid-Liquid Phase Separation for Poorly Water-Soluble Drugs. Mol Pharm 2024; 21:2878-2893. [PMID: 38767457 DOI: 10.1021/acs.molpharmaceut.4c00033] [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: 05/22/2024]
Abstract
Understanding the interplay between kinetics and thermodynamics of polymer-mediated liquid-liquid phase separation is crucial for designing and implementing an amorphous solid dispersion formulation strategy for poorly water-soluble drugs. This work investigates the phase behaviors of a poorly water-soluble model drug, celecoxib (CXB), in a supersaturated aqueous solution with and without polymeric additives (PVP, PVPVA, HPMCAS, and HPMCP). Drug-polymer-water ternary phase diagrams were also constructed to estimate the thermodynamic behaviors of the mixtures at room temperature. The liquid-liquid phase separation onset point for CXB was detected using an inline UV/vis spectrometer equipped with a fiber optic probe. Varying CXB concentrations were achieved using an accurate syringe pump throughout this study. The appearance of the transient nanodroplets was verified by cryo-EM and total internal reflection fluoresence microscopic techniques. The impacts of various factors, such as polymer composition, drug stock solution pumping rates, and the types of drug-polymer interactions, are tested against the onset points of the CXB liquid-liquid phase separation (LLPS). It was found that the types of drug-polymer interactions, i.e., hydrogen bonding and hydrophobic interactions, are vital to the position and shapes of LLPS in the supersaturation drug solution. A relation between the behaviors of LLPS and its location in the CXB-polymer-water ternary phase diagram was drawn from the findings.
Collapse
Affiliation(s)
- Kaijie Qian
- School of Pharmacy, McClay Research Centre, Queen's University Belfast, 97 Lisburn Road, Northern Ireland BT9 7BL, U.K
| | - Lorenzo Stella
- School of Mathematics and Physics, Queen's University Belfast, University Road, Belfast BT7 1NN, U.K
- School of Chemistry and Chemical Engineering, Queen's University Belfast, Stranmillis Road, Belfast BT9 5AG, U.K
| | - Fanjun Liu
- School of Pharmacy, McClay Research Centre, Queen's University Belfast, 97 Lisburn Road, Northern Ireland BT9 7BL, U.K
| | - David S Jones
- School of Pharmacy, McClay Research Centre, Queen's University Belfast, 97 Lisburn Road, Northern Ireland BT9 7BL, U.K
| | - Gavin P Andrews
- School of Pharmacy, McClay Research Centre, Queen's University Belfast, 97 Lisburn Road, Northern Ireland BT9 7BL, U.K
| | - Yiwei Tian
- School of Pharmacy, McClay Research Centre, Queen's University Belfast, 97 Lisburn Road, Northern Ireland BT9 7BL, U.K
| |
Collapse
|
12
|
Jain KMH, Hou HH, Siegel RA. An Artificial Gut/Absorption Simulator: Understanding the Impact of Absorption on In Vitro Dissolution, Speciation, and Precipitation of Amorphous Solid Dispersions. Mol Pharm 2024; 21:1884-1899. [PMID: 38512389 DOI: 10.1021/acs.molpharmaceut.3c01180] [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: 03/23/2024]
Abstract
Upon dissolution, amorphous solid dispersions (ASDs) of poorly water-soluble compounds can generate supersaturated solutions consisting of bound and free drug species that are in dynamic equilibrium with each other. Only free drug is available for absorption. Drug species bound to bile micelles, polymer excipients, and amorphous and crystalline precipitate can reduce the drug solute's activity to permeate, but they can also serve as reservoirs to replenish free drug in solution lost to absorption. However, with multiple processes of dissolution, absorption, and speciation occurring simultaneously, it may become challenging to understand which processes lead to an increase or decrease in drug solution concentration. Closed, nonsink dissolution testing methods used routinely, in the absence of drug removal, allow only for static equilibrium to exist and obscure the impact of each drug species on absorption. An artificial gut simulator (AGS) introduced recently consists of a hollow fiber-based absorption module and allows mass transfer of the drug from the dissolution media at a physiological rate after tuning the operating parameters. In the present work, ASDs of varying drug loadings were prepared with a BCS-II model compound, ketoconazole (KTZ), and hypromellose acetate succinate (HPMCAS) polymer. Simultaneous dissolution and absorption testing of the ASDs was conducted with the AGS, and simple analytical techniques were utilized to elucidate the impact of bound drug species on absorption. In all cases, a lower amount of crystalline precipitate was formed in the presence of absorption relative to the nonsink dissolution "control". However, formation of HPMCAS-bound drug species and crystalline precipitate significantly reduced KTZ absorption. Moreover, at high drug loading, inclusion of an absorption module was shown to enhance ASD dissolution. The rank ordering of the ASDs with respect to dissolution was significantly different when nonsink dissolution versus AGS was used, and this discrepancy could be mechanistically elucidated by understanding drug dissolution and speciation in the presence of absorption.
Collapse
Affiliation(s)
| | - Hao Helen Hou
- Small Molecule Pharmaceutical Sciences, Genentech Inc., South San Francisco, California 94080, United States
| | - Ronald A Siegel
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
13
|
Yoshikawa E, Ueda K, Hakata R, Higashi K, Moribe K. Quantitative Investigation of Intestinal Drug Absorption Enhancement by Drug-Rich Nanodroplets Generated via Liquid-Liquid Phase Separation. Mol Pharm 2024; 21:1745-1755. [PMID: 38501717 DOI: 10.1021/acs.molpharmaceut.3c01078] [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: 03/20/2024]
Abstract
Drug-rich droplets formed through liquid-liquid phase separation (LLPS) have the potential to enhance the oral absorption of drugs. This can be attributed to the diffusion of these droplets into the unstirred water layer (UWL) of the gastrointestinal tract and their reservoir effects on maintaining drug supersaturation. However, a quantitative understanding of the effect of drug-rich droplets on intestinal drug absorption is still lacking. In this study, the enhancement of intestinal drug absorption through the formation of drug-rich droplets was quantitatively evaluated on a mechanistic basis. To obtain fenofibrate (FFB)-rich droplets, an amorphous solid dispersion (ASD) of FFB/hypromellose (HPMC) was dispersed in an aqueous medium. Physicochemical characterization confirmed the presence of nanosized FFB-rich droplets in the supercooled liquid state within the FFB/HPMC ASD dispersion. An in situ single-pass intestinal perfusion (SPIP) assay in rats demonstrated that increased quantities of FFB-rich nanodroplets enhanced the intestinal absorption of FFB. The effective diffusion of FFB-rich nanodroplets through UWL would partially contribute to the improved FFB absorption. Additionally, confocal laser scanning microscopy (CLSM) of cross sections of the rat intestine after the administration of fluorescently labeled FFB-rich nanodroplets showed that these nanodroplets were directly taken up by small intestinal epithelial cells. Therefore, the direct uptake of drug-rich nanodroplets by the small intestine is a potential mechanism for improving FFB absorption in the intestine. To quantitatively evaluate the impact of FFB-rich droplets on the FFB absorption enhancement, we determined the apparent permeabilities of the FFB-rich nanodroplets and dissolved FFB based on the SPIP results. The apparent permeability of the FFB-rich nanodroplets was 110-130 times lower than that of dissolved FFB. However, when the FFB-rich nanodroplet concentration was several hundred times higher than that of dissolved FFB, the FFB-rich nanodroplets contributed significantly to FFB absorption improvement. The present study highlights that drug-rich nanodroplets play a direct role in enhancing drug absorption in the gastrointestinal tract, indicating their potential for further improvement of oral absorption from ASD formulations.
Collapse
Affiliation(s)
- Etsushi Yoshikawa
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Keisuke Ueda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Rei Hakata
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Kenjirou Higashi
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Kunikazu Moribe
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| |
Collapse
|
14
|
Sinko PD, Parker L, Prahl Wittberg L, Bergström CAS. Estimation of the concentration boundary layer adjacent to a flat surface using computational fluid dynamics. Int J Pharm 2024; 653:123870. [PMID: 38401511 DOI: 10.1016/j.ijpharm.2024.123870] [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: 08/10/2023] [Revised: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 02/26/2024]
Abstract
Dissolution-permeation (D/P) experiments are widely used during preclinical development due to producing results with better predictability than traditional monophasic experiments. However, it is difficult to compare absorption across in vitro setups given the propensity to only report apparent permeability. We therefore developed an approach to predict the concentration boundary layer for any D/P device by using computational fluid dynamics (CFD). The Navier-Stokes and continuity equation in 2D were solved numerically in MATLAB and by finite element methods in COMSOL v6.1 to predict the momentum [Formula: see text] and concentration ηg boundary layer for a flow over a flat plate, i.e. the classical Blasius boundary layer flow. A MATLAB algorithm was developed to calculate the edge of either boundary layer. The methodology to determine the concentration boundary layer based on Blasius's analysis provided an accurate estimate for both [Formula: see text] and ηg, resulting in, [Formula: see text] , at high Schmidt numbers (Sc ∼ 1000) within 14 % of the Blasius solution and 6.6 % of the accepted Schmidt number correlation ( [Formula: see text] ). The methodology based on the Blasius analysis of the concentration boundary layer using velocity and concentration profiles computed using CFD presented herein will enable characterization/analysis of complex D/P apparatuses used in preclinical development, where an analytical solution may not be available.
Collapse
Affiliation(s)
- Patrick D Sinko
- Department of Pharmacy, Uppsala Biomedical Center, Uppsala University, 751 23 Uppsala, Sweden
| | - Louis Parker
- FLOW, Department of Engineering Mechanics, Royal Institute of Technology, KTH, Osquars Backe 18, SE-100 44 Stockholm, Sweden
| | - Lisa Prahl Wittberg
- FLOW, Department of Engineering Mechanics, Royal Institute of Technology, KTH, Osquars Backe 18, SE-100 44 Stockholm, Sweden
| | - Christel A S Bergström
- Department of Pharmacy, Uppsala Biomedical Center, Uppsala University, 751 23 Uppsala, Sweden.
| |
Collapse
|
15
|
Chen G, Zhu Y, Wang Q, Bai Y, Ma S, Wang J, Zhao M, Zou M, Cheng G. The development of a novel simultaneous in vitro dissolution - in situ perfusion system as a potential tool for studying the absorption of solid oral formulation in rat. Eur J Pharm Sci 2023; 191:106601. [PMID: 37783379 DOI: 10.1016/j.ejps.2023.106601] [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: 05/10/2023] [Revised: 09/03/2023] [Accepted: 09/29/2023] [Indexed: 10/04/2023]
Abstract
The aim of this work is to develop a novel simultaneous in vitro dissolution - in situ perfusion system (SDPS) as a potential tool to evaluate the in vivo performance of solid oral formulation in rat. The innovative nitrendipine (NTD) tablet of Bayotensin mite® made in Germany was used as reference listed drug (RLD), and five generic products from Chinese market were compared with RLD using the in vitro dissolution test method specified by the orange book and the SDPS method developed in this study. Four self-prepared NTD tablets with different proportions of microcrystalline cellulose/starch were employed to investigate the discriminatory ability of the SDPS for formulation. In addition, the predictivity of the SDPS in relation to data from in vivo pharmaceutics studies was evaluated. The 45-min dissolution test and multiple-pH dissolution profiles of generic product 1 and 2 have no difference compared with the RLD, but their dissolution profiles from the SDPS showed statistically significant differences. A biexponential formula successfully described the concentration profiles of self-prepared formulations in SDPS experiments. The kdis (0.08 ± 0.01 ∼ 0.2 ± 0.03 min-1) and ka (about 2.30 × 10-3 min-1) values calculated by the formulas of F1-F3 suggested that the used excipients had no effect on the intestinal absorption of NTD, and it might be the property of active pharmaceutical ingredient that led to the difference among the generics. Furthermore, the in vivo rat pharmacokinetics study results of F1-F3 showed a good correlation (R2 = 0.99) with the SDPS data. In summary, the SDPS is a promising tool to detect the unexpected quality changes of pharmaceutical products in weakly regulated markets, facilitate formulation screening, and potentially reduce animal testing for estimating the in vivo absorption behavior of solid oral formulations. The absorption performance of generic drugs in vivo should be further investigated.
Collapse
Affiliation(s)
- Guo Chen
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang 110016, China
| | - Yumeng Zhu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang 110016, China
| | - Qiaoqiao Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang 110016, China
| | - Yifeng Bai
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang 110016, China
| | - Siyuan Ma
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang 110016, China
| | - Jingfeng Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang 110016, China
| | - Minqian Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang 110016, China
| | - Meijuan Zou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang 110016, China
| | - Gang Cheng
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, No.103, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
16
|
Moseson DE, Taylor LS. Crystallinity: A Complex Critical Quality Attribute of Amorphous Solid Dispersions. Mol Pharm 2023; 20:4802-4825. [PMID: 37699354 DOI: 10.1021/acs.molpharmaceut.3c00526] [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: 09/14/2023]
Abstract
Does the performance of an amorphous solid dispersion rely on having 100% amorphous content? What specifications are appropriate for crystalline content within an amorphous solid dispersion (ASD) drug product? In this Perspective, the origin and significance of crystallinity within amorphous solid dispersions will be considered. Crystallinity can be found within an ASD from one of two pathways: (1) incomplete amorphization, or (2) crystal creation (nucleation and crystal growth). While nucleation and crystal growth is the more commonly considered pathway, where crystals originate as a physical stability failure upon accelerated or prolonged storage, manufacturing-based origins of crystallinity are possible as well. Detecting trace levels of crystallinity is a significant analytical challenge, and orthogonal methods should be employed to develop a holistic assessment of sample properties. Probing the impact of crystallinity on release performance which may translate to meaningful clinical significance is inherently challenging, requiring optimization of dissolution test variables to address the complexity of ASD formulations, in terms of drug physicochemical properties (e.g., crystallization tendency), level of crystallinity, crystal reference material selection, and formulation characteristics. The complexity of risk presented by crystallinity to product performance will be illuminated through several case studies, highlighting that a one-size-fits-all approach cannot be used to set specification limits, as the risk of crystallinity can vary widely based on a multitude of factors. Risk assessment considerations surrounding drug physicochemical properties, formulation fundamentals, physical stability, dissolution, and crystal micromeritic properties will be discussed.
Collapse
Affiliation(s)
- Dana E Moseson
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
- Worldwide Research and Development Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
17
|
Supersaturation and phase behavior during dissolution of amorphous solid dispersions. Int J Pharm 2023; 631:122524. [PMID: 36549404 DOI: 10.1016/j.ijpharm.2022.122524] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/04/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022]
Abstract
Amorphous solid dispersion (ASD) is a promising strategy to enhance solubility and bioavailability of poorly water-soluble drugs. Due to higher free energy of ASD, supersaturated drug solution could be generated during dissolution. When amorphous solubility of a drug is exceeded, drug-rich nanodroplets could form and act as a reservoir to maintain the maximum free drug concentration in solution, facilitating the absorption of the drug in vivo. Dissolution behavior of ASD has received increasing interests. This review will focus on the recent advances in ASD dissolution, including the generation and maintenance of supersaturated drug solution in absence or presence of liquid-liquid phase separation. Mechanism of drug release from ASD including polymer-controlled dissolution and drug-controlled dissolution will be introduced. Formation of amorphous drug-rich nanodroplets during dissolution and the underlying mechanism will be discussed. Phase separation morphology of hydrated ASD plays a critical role in dissolution behavior of ASD, which will be highlighted. Supersaturated drug solution shows poor physical stability and tends to crystallize. The effect of polymer and surfactant on supersaturated drug solution will be demonstrated and some unexpected results will be shown. Physicochemical properties of drug and polymer could impact ASD dissolution and some of them even show opposite effect on dissolution and physical stability of ASD in solid state, respectively. This review will contribute to a better understanding of ASD dissolution and facilitate a rational design of ASD formulation.
Collapse
|
18
|
Pöstges F, Kayser K, Appelhaus J, Monschke M, Gütschow M, Steinebach C, Wagner KG. Solubility Enhanced Formulation Approaches to Overcome Oral Delivery Obstacles of PROTACs. Pharmaceutics 2023; 15:pharmaceutics15010156. [PMID: 36678785 PMCID: PMC9863516 DOI: 10.3390/pharmaceutics15010156] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/23/2022] [Accepted: 12/30/2022] [Indexed: 01/04/2023] Open
Abstract
PROteolysis TArgeting Chimaeras (PROTACs) offer new opportunities in modern medicine by targeting proteins that are undruggable to classic inhibitors. However, due to their hydrophobic structure, PROTACs typically suffer from low solubility, and oral bioavailability remains challenging. At the same time, due to their investigative state, the drug supply is meager, leading to limited possibilities in terms of formulation development. Therefore, we investigated the solubility enhancement employing mini-scale formulations of amorphous solid dispersions (ASDs) and liquisolid formulations of the prototypic PROTAC ARCC-4. Based on preliminary supersaturation testing, HPMCAS (L Grade) and Eudragit® L 100-55 (EL 100-55) were demonstrated to be suitable polymers for supersaturation stabilization of ARCC-4. These two polymers were selected for preparing ASDs via vacuum compression molding (VCM), using drug loads of 10 and 20%, respectively. The ASDs were subsequently characterized with respect to their solid state via differential scanning calorimetry (DSC). Non-sink dissolution testing revealed that the physical mixtures (PMs) did not improve dissolution. At the same time, all ASDs enabled pronounced supersaturation of ARCC-4 without precipitation for the entire dissolution period. In contrast, liquisolid formulations failed in increasing ARCC-4 solubility. Hence, we demonstrated that ASD formation is a promising principle to overcome the low solubility of PROTACs.
Collapse
Affiliation(s)
- Florian Pöstges
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Bonn, Gerhard-Domagk-Str. 3, 53121 Bonn, Germany
| | - Kevin Kayser
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Bonn, Gerhard-Domagk-Str. 3, 53121 Bonn, Germany
| | - Jan Appelhaus
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Bonn, Gerhard-Domagk-Str. 3, 53121 Bonn, Germany
| | - Marius Monschke
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Bonn, Gerhard-Domagk-Str. 3, 53121 Bonn, Germany
| | - Michael Gütschow
- Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Christian Steinebach
- Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
- Correspondence: (C.S.); (K.G.W.); Tel.: +49-228-73-2308 (C.S.); +49-228-73-5271 (K.G.W.)
| | - Karl G. Wagner
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Bonn, Gerhard-Domagk-Str. 3, 53121 Bonn, Germany
- Correspondence: (C.S.); (K.G.W.); Tel.: +49-228-73-2308 (C.S.); +49-228-73-5271 (K.G.W.)
| |
Collapse
|
19
|
Carvedilol Precipitation Inhibition by the Incorporation of Polymeric Precipitation Inhibitors Using a Stable Amorphous Solid Dispersion Approach: Formulation, Characterization, and In Vitro In Vivo Evaluation. Polymers (Basel) 2022; 14:polym14224977. [PMID: 36433104 PMCID: PMC9697141 DOI: 10.3390/polym14224977] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/05/2022] [Accepted: 11/10/2022] [Indexed: 11/19/2022] Open
Abstract
An amorphous solid dispersion (ASD) of carvedilol (CVL) was prepared via the solvent evaporation method, using cellulose derivatives as polymeric precipitation inhibitors (PPIs). The prepared ASDs existed in the amorphous phase, as revealed by X-ray powder diffraction (XRPD) and scanning electron microscopy (SEM). The Fourier-transform infrared spectroscopy (FT-IR) and differential scanning calorimetry (DSC) results confirmed the compatibility between CVL and the polymers used. The ASDs characteristics were evaluated, with no change in viscosity, a pH of 6.8, a polydispersity index of 0.169, a particle size of 423-450 nm, and a zeta potential of 3.80 mV. Crystal growth inhibition was assessed for 180 min via an infusion precipitation study in simulated intestinal fluid (SIF). The interactions between the drug and polymers were established in great detail, using nuclear magnetic resonance (NMR) spectroscopy, nuclear Overhauser effect spectroscopy (NOESY), and Raman spectroscopy studies. Dielectric analysis was employed to determine the drug-polymer interactions between ion pairs and to understand ion transport behavior. In vivo oral kinetics and irritation studies performed on Wistar rats have demonstrated promising biocompatibility, stability, and the enhanced bioavailability of CVL. Collectively, the stable ASDs of CVL were developed using cellulose polymers as PPIs that would inhibit drug precipitation in the gastrointestinal tract and would aid in achieving higher in vivo drug stability and bioavailability.
Collapse
|
20
|
Sabra R, Narula A, Taylor LS, Li N. Comparisons of in Vitro Models to Evaluate the Membrane Permeability of Amorphous Drug Nanoparticles. Mol Pharm 2022; 19:3412-3428. [PMID: 35972995 DOI: 10.1021/acs.molpharmaceut.2c00565] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The spontaneous formation of amorphous drug nanoparticles following the release of a drug from a supersaturating formulation is gaining increasing attention due to their potential contribution to increased oral bioavailability. The formation of nanosized drug particles also has considerable implications for the interpretation of in vitro and in vivo data. However, the membrane transport properties of these drug particles remain less well understood. Herein, the membrane permeation of nanosized amorphous drug particles of a model drug atazanavir was evaluated using different artificial membrane-based, cell-based, and animal tissue-based models. Results showed that flux enhancement by particles was different for the various systems used. Generally, good agreement was obtained among experiments performed using the same apparatus with different model membranes, with the exception of the Madin-Darby canine kidney cell monolayer and the Long-Evans rat intestine tissue, which showed lower flux enhancements. Franz cell-based models showed slightly higher flux enhancements by particles compared to Transwell and intestinal tissue sac models. Mass transport analysis suggested that the extent of flux enhancement by particles is dependent on the geometry of the apparatus as well as the properties of the membrane and buffer used, whereas the flux plateau concentration is dependent on the unstirred water later (UWL) asymmetry. These results highlight the complexity in characterizing the permeability advantage of these nonmembrane permeable drug particles and suggest that caution should be used in selecting the appropriate in vitro model to evaluate the overall permeability of colloidal drug particles.
Collapse
Affiliation(s)
- Rayan Sabra
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269, United States
| | - Akshay Narula
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Na Li
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269, United States.,Institute of Materials Science, University of Connecticut, 97 North Eagleville Road, Unit 3136, Storrs, Connecticut 06269, United States
| |
Collapse
|
21
|
Microdialysis and nanofiltration allow to distinguish molecularly dissolved from colloid- associated drug concentrations during biomimetic dissolution testing of supersaturating formulations. Eur J Pharm Sci 2022; 174:106166. [DOI: 10.1016/j.ejps.2022.106166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/14/2022] [Accepted: 03/08/2022] [Indexed: 11/23/2022]
|
22
|
Indulkar AS, Lou X, Zhang GGZ, Taylor LS. Role of Surfactants on Release Performance of Amorphous Solid Dispersions of Ritonavir and Copovidone. Pharm Res 2022; 39:381-397. [PMID: 35169959 DOI: 10.1007/s11095-022-03183-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/26/2022] [Indexed: 02/06/2023]
Abstract
PURPOSE To understand the role of different surfactants, incorporated into amorphous solid dispersions (ASDs) of ritonavir and copovidone, in terms of their impact on release, phase behavior and stabilization of amorphous precipitates formed following drug release. METHODS Ternary ASDs with ritonavir, copovidone and surfactants (30:70:5 w/w/w) were prepared by rotary evaporation. ASD release performance was tested using Wood's intrinsic dissolution rate apparatus and compared to the binary drug-polymer ASD with 30% drug loading. Size measurement of amorphous droplets was performed using dynamic light scattering. Solid state characterization was performed using attenuated total reflectance-infrared spectroscopy, differential scanning calorimetry and scanning electron microscopy. RESULTS All surfactant-containing ASDs showed improvement over the binary ASD. Span 85 and D-α-tocopheryl polyethylene glycol succinate (TPGS) showed complete release with no evidence of AAPS or crystallization whereas Span 20 and Tween 80 showed < 50% release with amorphous amorphous phase separation (AAPS). Span 20 also induced solution crystallization. Sodium dodecyl sulfate (SDS) showed very rapid, albeit incomplete (~ 80%) release. AAPS was not observed with SDS. However, crystallization on the dissolving solid surface was noted. Span 20 and TPGS formed the smallest and most size-stable droplets with ~ 1 µm size whereas coalescence was noted with other surfactants. CONCLUSIONS Surfactants improved the release performance relative to the binary ASD. Different surfactant types impacted overall performance to varying extents and affected different attributes. Overall, Span 85 showed best performance (complete release, no crystallization/AAPS and small droplet size). Correlation between physicochemical properties and surfactant performance was not observed.
Collapse
Affiliation(s)
- Anura S Indulkar
- Drug Product Development, Research and Development, AbbVie Inc., N Waukegan Road, North Chicago, IL, 60064, USA
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN, 47907, USA
| | - Xiaochun Lou
- Drug Product Development, Research and Development, AbbVie Inc., N Waukegan Road, North Chicago, IL, 60064, USA
| | - Geoff G Z Zhang
- Drug Product Development, Research and Development, AbbVie Inc., N Waukegan Road, North Chicago, IL, 60064, USA.
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN, 47907, USA.
| |
Collapse
|
23
|
Lentz KA, Plum J, Steffansen B, Arvidsson PO, Omkvist DH, Pedersen AJ, Sennbro CJ, Pedersen GP, Jacobsen J. Predicting in vivo performance of fenofibrate amorphous solid dispersions using in vitro non-sink dissolution and dissolution permeation setup. Int J Pharm 2021; 610:121174. [PMID: 34655705 DOI: 10.1016/j.ijpharm.2021.121174] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/01/2021] [Accepted: 10/04/2021] [Indexed: 11/17/2022]
Abstract
Amorphous solid dispersion (ASD) is emerging as a useful formulation strategy to increase the bioavailability of active pharmaceutical ingredients with poor solubility. In vitro dissolution testing under non-sink conditions has often been used to evaluate the ability of ASDs to generate and maintain supersaturation to predict the in vivo performance. However, such a single compartment dissolution setup can fail to predict the oral bioavailability, due to an interdependence between precipitation and permeation. Hence, the use of two compartment dissolution-permeation setups is emerging. In this study, three ASDs containing fenofibrate as model drug substance were developed using Soluplus®, and Hypromellose Acetate Succinate in two different grades (high and low), respectively. The aim was to compare the use of a small-scale in vitro non-sink dissolution setup and a small-scale in vitro dissolution-permeation setup to predict the in vivo oral exposure of the ASDs in rats. The maximum concentration (Cmax) and area under curve (AUC) obtained in the in vitro studies were used to predict the in vivo rank order of the formulations. The results showed that the two in vitro studies resulted in the same rank order based on both Cmax and AUC. Interestingly, Cmax resulted in a better in vitro/in vivo correlation than the in vitro AUC, and based on the in vitro Cmax, the in vivo rank order was predicted.
Collapse
Affiliation(s)
- Karoline Aagaard Lentz
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; LEO Pharma, A/S, Industriparken 55, DK-2750 Ballerup, Denmark
| | - Jakob Plum
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; LEO Pharma, A/S, Industriparken 55, DK-2750 Ballerup, Denmark.
| | | | | | | | | | | | | | - Jette Jacobsen
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| |
Collapse
|
24
|
El Sayed M, Alhalaweh A, Bergström CAS. Impact of Simulated Intestinal Fluids on Dissolution, Solution Chemistry, and Membrane Transport of Amorphous Multidrug Formulations. Mol Pharm 2021; 18:4079-4089. [PMID: 34613730 PMCID: PMC8564758 DOI: 10.1021/acs.molpharmaceut.1c00480] [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] [Indexed: 11/28/2022]
Abstract
![]()
The solution behavior
and membrane transport of multidrug formulations
were herein investigated in a biorelevant medium simulating fasted
conditions. Amorphous multidrug formulations were prepared by the
solvent evaporation method. Combinations of atazanavir (ATV) and ritonavir
(RTV) and felodipine (FDN) and indapamide (IPM) were prepared and
stabilized by a polymer for studying their dissolution (under non-sink
conditions) and membrane transport in fasted state simulated intestinal
fluid (FaSSIF). The micellar solubilization by FaSSIF enhanced the
amorphous solubility of the drugs to different extents. Similar to
buffer, the maximum achievable concentration of drugs in combination
was reduced in FaSSIF, but the extent of reduction was affected by
the degree of FaSSIF solubilization. Dissolution studies of ATV and
IPM revealed that the amorphous solubility of these two drugs was
not affected by FaSSIF solubilization. In contrast, RTV was significantly
affected by FaSSIF solubilization with a 30% reduction in the maximum
achievable concentration upon combination to ATV, compared to 50%
reduction in buffer. This positive deviation by FaSSIF solubilization
was not reflected in the mass transport–time profiles. Interestingly,
FDN concentrations remain constant until the amount of IPM added was
over 1000 μg/mL. No decrease in the membrane transport of FDN
was observed for a 1:1 M ratio of FDN-IPM combination. This study
demonstrates the importance of studying amorphous multidrug formulations
under physiologically relevant conditions to obtain insights into
the performance of these formulations after oral administration.
Collapse
Affiliation(s)
- Mira El Sayed
- Department of Pharmacy, Biomedical Centre, Uppsala University, P.O. Box 580, Uppsala SE-751 23, Sweden.,Recipharm OT Chemistry AB, Uppsala SE-754 50, Sweden
| | | | - Christel A S Bergström
- Department of Pharmacy, Biomedical Centre, Uppsala University, P.O. Box 580, Uppsala SE-751 23, Sweden
| |
Collapse
|
25
|
Ramachandran G, Sudheesh MS. Role of Permeability on the Biopredictive Dissolution of Amorphous Solid Dispersions. AAPS PharmSciTech 2021; 22:243. [PMID: 34595565 DOI: 10.1208/s12249-021-02125-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 08/23/2021] [Indexed: 02/08/2023] Open
Abstract
An ideal dissolution test for amorphous solid dispersions (ASDs) should reflect physicochemical, physiological, and hydrodynamic conditions which accurately represent in vivo dissolution. However, this is confounded by the evolution of different molecular and colloidal species during dissolution, generating a supersaturated state of the drug. The supersaturated state of a drug is thermodynamically unstable which drives the process of precipitation resulting in a loss of solubility advantage. Maintaining a supersaturated state of the drug with the help of precipitation inhibiting excipients is a key component in the design of ASDs. Therefore, a biopredictive dissolution test is critical for proper risk assessment during the development of an optimal ASD formulation. One of the overlooked components of biopredictive dissolution is the role of drug permeability. The kinetic changes in the phase behavior of a drug during dissolution of ASDs are influenced by drug permeability across a membrane. Conventionally, drug dissolution and permeation are analyzed separately although they occur simultaneously in vivo. The kinetic phase changes occurring during dissolution of ASDs can influence the thermodynamic activity and membrane flux of a drug. The present review evaluates the feasibility, predictability, and practicability of permeability/dissolution for the optimal development and risk assessment of ASD formulations.
Collapse
|
26
|
Hate SS, Mosquera-Giraldo LI, Taylor LS. A Mechanistic Study of Drug Mass Transport from Supersaturated Solutions Across PAMPA Membranes. J Pharm Sci 2021; 111:102-115. [PMID: 34237298 DOI: 10.1016/j.xphs.2021.07.003] [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: 04/27/2021] [Revised: 06/30/2021] [Accepted: 07/02/2021] [Indexed: 10/20/2022]
Abstract
There is an increasing shift from dissolution testing to dissolution-permeation testing of formulations during formulation development and this has led increasing application of permeability measurements using parallel artificial membrane permeability assay (PAMPA) membranes. However, there is a lack of thorough analysis of the impact of variabilities in the PAMPA setup on the mass flow rate outcomes, particularly for complex solubility-enabling formulations. In this study, we investigated the impact of amorphous drug-rich nanodroplets, formed in supersaturated solutions by liquid-liquid phase separation, on membrane transport by measuring mass flow rate across PAMPA membranes. In addition, we explored the impact of PAMPA variants such as lipid composition, hydrophobicity and pore size of the filter support, as well as receiver sink properties on membrane mass flow rates of solutions containing amorphous nanodroplets. Filter properties and lipid composition did not show a notable influence on the mass flow rates for lipophilic molecules, while a marked impact was observed for hydrophilic molecules. High sink conditions in the receiver compartment, arising from addition of micellar surfactant, altered the membrane integrity for lipid-impregnated hydrophilic membranes. In contrast, no such effect was observed for a hydrophobic filter support. Membrane integrity tests also suggested that monitoring water transport may be an improved approach over using Lucifer yellow. Furthermore, high sink conditions in the receiver compartment resulted in an increase in the overall mass flow rate. This was due to the effect of asymmetric conditions, generated across the membrane, on mass transport kinetics. Linearity between mass flow rate and donor concentration was observed until the donor concentration reached the amorphous solubility. Above the amorphous solubility, a gradual increase in mass flow rate was observed i.e., with an increasing number of nanodroplets in the solution. This was attributed to decrease in the permeability barrier across unstirred water layer due to reduction of the concentration gradient as nanodroplets dissolved to replenish absorbed drug. Observations made in this study provide insights into the mechanisms associated with mass transport of supersaturated solutions across PAMPA membranes, which are critical for improved evaluation of enabling formulations.
Collapse
Affiliation(s)
- Siddhi S Hate
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN, United States
| | - Laura I Mosquera-Giraldo
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN, United States.
| |
Collapse
|
27
|
Qian K, Stella L, Jones DS, Andrews GP, Du H, Tian Y. Drug-Rich Phases Induced by Amorphous Solid Dispersion: Arbitrary or Intentional Goal in Oral Drug Delivery? Pharmaceutics 2021; 13:889. [PMID: 34203969 PMCID: PMC8232734 DOI: 10.3390/pharmaceutics13060889] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 12/19/2022] Open
Abstract
Among many methods to mitigate the solubility limitations of drug compounds, amorphous solid dispersion (ASD) is considered to be one of the most promising strategies to enhance the dissolution and bioavailability of poorly water-soluble drugs. The enhancement of ASD in the oral absorption of drugs has been mainly attributed to the high apparent drug solubility during the dissolution. In the last decade, with the implementations of new knowledge and advanced analytical techniques, a drug-rich transient metastable phase was frequently highlighted within the supersaturation stage of the ASD dissolution. The extended drug absorption and bioavailability enhancement may be attributed to the metastability of such drug-rich phases. In this paper, we have reviewed (i) the possible theory behind the formation and stabilization of such metastable drug-rich phases, with a focus on non-classical nucleation; (ii) the additional benefits of the ASD-induced drug-rich phases for bioavailability enhancements. It is envisaged that a greater understanding of the non-classical nucleation theory and its application on the ASD design might accelerate the drug product development process in the future.
Collapse
Affiliation(s)
- Kaijie Qian
- Pharmaceutical Engineering Group, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (K.Q.); (D.S.J.); (G.P.A.)
| | - Lorenzo Stella
- Atomistic Simulation Centre, School of Mathematics and Physics, Queen’s University Belfast, 7–9 College Park E, Belfast BT7 1PS, UK;
- David Keir Building, School of Chemistry and Chemical Engineering, Queen’s University Belfast, Stranmillis Road, Belfast BT9 5AG, UK
| | - David S. Jones
- Pharmaceutical Engineering Group, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (K.Q.); (D.S.J.); (G.P.A.)
| | - Gavin P. Andrews
- Pharmaceutical Engineering Group, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (K.Q.); (D.S.J.); (G.P.A.)
- School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Huachuan Du
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
- Simpson Querrey Institute, Northwestern University, 303 East Superior Street, 11th floor, Chicago, IL 60611, USA
| | - Yiwei Tian
- Pharmaceutical Engineering Group, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (K.Q.); (D.S.J.); (G.P.A.)
| |
Collapse
|
28
|
Vinarov Z, Abrahamsson B, Artursson P, Batchelor H, Berben P, Bernkop-Schnürch A, Butler J, Ceulemans J, Davies N, Dupont D, Flaten GE, Fotaki N, Griffin BT, Jannin V, Keemink J, Kesisoglou F, Koziolek M, Kuentz M, Mackie A, Meléndez-Martínez AJ, McAllister M, Müllertz A, O'Driscoll CM, Parrott N, Paszkowska J, Pavek P, Porter CJH, Reppas C, Stillhart C, Sugano K, Toader E, Valentová K, Vertzoni M, De Wildt SN, Wilson CG, Augustijns P. Current challenges and future perspectives in oral absorption research: An opinion of the UNGAP network. Adv Drug Deliv Rev 2021; 171:289-331. [PMID: 33610694 DOI: 10.1016/j.addr.2021.02.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/12/2021] [Accepted: 02/01/2021] [Indexed: 02/06/2023]
Abstract
Although oral drug delivery is the preferred administration route and has been used for centuries, modern drug discovery and development pipelines challenge conventional formulation approaches and highlight the insufficient mechanistic understanding of processes critical to oral drug absorption. This review presents the opinion of UNGAP scientists on four key themes across the oral absorption landscape: (1) specific patient populations, (2) regional differences in the gastrointestinal tract, (3) advanced formulations and (4) food-drug interactions. The differences of oral absorption in pediatric and geriatric populations, the specific issues in colonic absorption, the formulation approaches for poorly water-soluble (small molecules) and poorly permeable (peptides, RNA etc.) drugs, as well as the vast realm of food effects, are some of the topics discussed in detail. The identified controversies and gaps in the current understanding of gastrointestinal absorption-related processes are used to create a roadmap for the future of oral drug absorption research.
Collapse
Affiliation(s)
- Zahari Vinarov
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium; Department of Chemical and Pharmaceutical Engineering, Sofia University, Sofia, Bulgaria
| | - Bertil Abrahamsson
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, Sweden
| | - Per Artursson
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Hannah Batchelor
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Philippe Berben
- Pharmaceutical Development, UCB Pharma SA, Braine- l'Alleud, Belgium
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - James Butler
- GlaxoSmithKline Research and Development, Ware, United Kingdom
| | | | - Nigel Davies
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Gøril Eide Flaten
- Department of Pharmacy, UiT The Arctic University of Norway, Tromsø, Norway
| | - Nikoletta Fotaki
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | | | | | | | | | | | - Martin Kuentz
- Institute for Pharma Technology, University of Applied Sciences and Arts Northwestern Switzerland, Basel, Switzerland
| | - Alan Mackie
- School of Food Science & Nutrition, University of Leeds, Leeds, United Kingdom
| | | | | | - Anette Müllertz
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Petr Pavek
- Faculty of Pharmacy, Charles University, Hradec Králové, Czech Republic
| | | | - Christos Reppas
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Kiyohiko Sugano
- College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan
| | - Elena Toader
- Faculty of Medicine, University of Medicine and Pharmacy of Iasi, Romania
| | - Kateřina Valentová
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Maria Vertzoni
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Saskia N De Wildt
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Clive G Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Patrick Augustijns
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
29
|
Ashwathy P, Anto AT, Sudheesh MS. A mechanistic review on the dissolution phase behavior and supersaturation stabilization of amorphous solid dispersions. Drug Dev Ind Pharm 2021; 47:1-11. [PMID: 33494623 DOI: 10.1080/03639045.2021.1879843] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Amorphous solid dispersion (ASD) technology is an attractive formulation approach for poorly soluble drugs because of the supersaturated state acquired during its dissolution. The high thermodynamic activity of the supersaturated state of the drug is also a driver for the enhanced absorptive flux across a membrane. However, this advantage can easily be lost due to the inherent instability of supersaturation, causing drug precipitation. Stabilizing the supersaturated state during the dissolution of ASD for the relevant absorption time frame is a challenging area in formulation research. Stabilizing the supersaturated state by using polymeric excipients and understanding the phase behavior of drugs during dissolution are required for the optimal performance of ASD formulations. A number of confounding kinetic, formulation and physiological factors can influence the evolution of supersaturation and phase changes during dissolution of ASDs. The review highlights the complex nature of dissolution of ASDs and the need of biorelevant dissolution for proper risk assessment and optimizing formulation development.
Collapse
Affiliation(s)
- P Ashwathy
- Department of Pharmaceutics, Amrita School of Pharmacy, Kochi, India
| | - Akshaya T Anto
- Department of Pharmaceutics, Amrita School of Pharmacy, Kochi, India
| | - M S Sudheesh
- Department of Pharmaceutics, Amrita School of Pharmacy, Kochi, India
| |
Collapse
|
30
|
Arce FA, Setiawan N, Campbell HR, Lu X, Nethercott MJ, Bummer P, Su Y, Marsac PJ. Toward Developing Discriminating Dissolution Methods for Formulations Containing Nanoparticulates in Solution: The Impact of Particle Drift and Drug Activity in Solution. Mol Pharm 2020; 17:4125-4140. [PMID: 32965123 DOI: 10.1021/acs.molpharmaceut.0c00599] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Enabling formulations are an attractive approach to increase the dissolution rate, solubility, and oral bioavailability of poorly soluble compounds. With the growing prevalence of poorly soluble drug compounds in the pharmaceutical pipeline, supersaturating drug delivery systems (SDDS), a subset of enabling formulations, have grown in popularity due to their properties allowing for drug concentrations greater than the corresponding crystalline solubility. However, the extent of supersaturation generated as the enabling formulation traverses the gastrointestinal (GI) tract is dynamic and poorly understood. The dynamic nature of supersaturation is a result of several competing kinetic processes such as dissolution, solubilization by formulation and endogenous surfactants, crystallization, and absorption. Ultimately, the free drug concentration, which is equivalent to the drug's inherent thermodynamic activity amid these kinetic processes, defines the true driving force for drug absorption. However, in cases where solubilizing agents are present (i.e., surfactants and bile salts), drug molecules may associate with colloidal nanoscale species, complicating drug activity determination. These nanoscale species can drift into the aqueous boundary layer (ABL), increasing the local API activity at the membrane surface, resulting in increased bioavailability. Herein, a novel approach was developed to accurately measure thermodynamic drug activity in complex media containing drug distributed in nanoparticulate species. This approach captures the influence of the ABL on the observed flux and, ultimately, the predicted unbound drug concentration. The results demonstrate that this approach can help to (1) measure the true extent of local supersaturation in complex systems containing solubilizing excipients and (2) elucidate the mechanisms by which colloidal aggregates can modulate the drug activity in solution and potentially enhance the flux observed across a membrane. The utilization of these techniques may provide development scientists with a strategy to evaluate formulation sensitivity to nanospeciation and allow formulators to maximize the driving force for absorption in a complex environment, perhaps enabling the development of dissolution methods with greater discrimination and correlation to pre-clinical and clinical data sets.
Collapse
Affiliation(s)
- Freddy A Arce
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
| | - Nico Setiawan
- Albany Molecular Research Inc., West Lafayette, Indiana 47906, United States
| | - Heather R Campbell
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
| | - Xingyu Lu
- Pharmaceutical Sciences, Merck & Co., Kenilworth, New Jersey 07033, United States.,Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, Hangzhou, Zhejiang 310024, China
| | | | - Paul Bummer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
| | - Yongchao Su
- Pharmaceutical Sciences, Merck & Co., Kenilworth, New Jersey 07033, United States.,Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States.,Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Patrick J Marsac
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
| |
Collapse
|
31
|
The tangential flow absorption model (TFAM) – A novel dissolution method for evaluating the performance of amorphous solid dispersions of poorly water-soluble actives. Eur J Pharm Biopharm 2020; 154:74-88. [DOI: 10.1016/j.ejpb.2020.06.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 05/01/2020] [Accepted: 06/19/2020] [Indexed: 11/19/2022]
|
32
|
Hate SS, Reutzel-Edens SM, Taylor LS. Influence of Drug-Silica Electrostatic Interactions on Drug Release from Mesoporous Silica-Based Oral Delivery Systems. Mol Pharm 2020; 17:3435-3446. [PMID: 32790416 DOI: 10.1021/acs.molpharmaceut.0c00488] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Mesoporous silica particles are attractive carriers for poorly soluble drugs whereby confinement of drugs in the mesopores leads to amorphization, which makes them potential carriers for enhanced oral delivery. However, interactions between the drug molecules and the silica surface can lead to incomplete drug release. The strength of the interaction depends on the silica surface chemistry, which varies as a function of pH, as well as on drug chemistry and ionization states. Herein, the adsorption and dissolution behavior of weakly basic drugs were evaluated as a function of pH to understand the impact of electrostatic interactions on the performance of mesoporous silica-based formulations. A higher adsorption was noted when the drug interacted with the silica surface via electrostatic interactions compared to hydrogen bonding. Higher adsorption, in turn, led to a lower extent of drug release. In two-stage release studies of drugs with pKa values close to the intestinal pH, a shift from low to higher pH solutions resulted in a decrease in the solution concentration. Further investigations demonstrated that this was due to readsorption of the drug, initially released in the acidic medium when the pH was increased. Two-stage release studies were also coupled with mass transport measurements. Only a slight improvement in drug release due to simultaneous absorption across a membrane was observed, suggesting strong drug adsorption to the silica surface arising from favorable electrostatic interactions, which diminishes the effect of sink conditions provided by the absorptive environment. This study highlights that physiological parameters, such as solution pH, are important considerations when designing mesoporous silica-based formulations for poorly soluble drugs. It also underscores the importance of incorporating in vivo-relevant conditions in in vitro testing to better evaluate these complex formulations due to the notable effect of dissolution media on the release behavior.
Collapse
Affiliation(s)
- Siddhi S Hate
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Susan M Reutzel-Edens
- Lilly Research Laboratories, Eli Lilly and Co., Indianapolis, Indiana 46285, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
33
|
Interplay of Adsorption, Supersaturation and the Presence of an Absorptive Sink on Drug Release from Mesoporous Silica-Based Formulations. Pharm Res 2020; 37:163. [DOI: 10.1007/s11095-020-02879-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/10/2020] [Indexed: 11/26/2022]
|
34
|
Ueda K, Hate SS, Taylor LS. Impact of Hypromellose Acetate Succinate Grade on Drug Amorphous Solubility and In Vitro Membrane Transport. J Pharm Sci 2020; 109:2464-2473. [DOI: 10.1016/j.xphs.2020.04.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/14/2020] [Accepted: 04/20/2020] [Indexed: 12/22/2022]
|
35
|
Impact of HPMCAS on the Dissolution Performance of Polyvinyl Alcohol Celecoxib Amorphous Solid Dispersions. Pharmaceutics 2020; 12:pharmaceutics12060541. [PMID: 32545270 PMCID: PMC7356348 DOI: 10.3390/pharmaceutics12060541] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 12/12/2022] Open
Abstract
Amorphous solid dispersions (ASDs) have been proven to increase the bioavailability of poorly soluble drugs. It is desirable that the ASD provide a rapid dissolution rate and a sufficient stabilization of the generated supersaturation. In many cases, one polymer alone is not able to provide both features, which raises a need for reasonable polymer combinations. In this study we aimed to generate a rapidly dissolving ASD using the hydrophilic polymer polyvinyl alcohol (PVA) combined with a suitable precipitation inhibitor. Initially, PVA and hydroxypropylmethylcellulose acetate succinate (HPMCAS) were screened for their precipitation inhibitory potential for celecoxib in solution. The generated supersaturation in presence of PVA or HPMCAS was further characterized using dynamic light scattering. Binary ASDs of either PVA or HPMCAS (at 10% and 20% drug load) were prepared by hot-melt extrusion and solid-state analytics were conducted using differential scanning calorimetry (DSC), X-ray powder diffraction (XRPD) and fourier-transformed infrared spectroscopy (FT-IR). The non-sink dissolution studies of the binary ASDs revealed a high dissolution rate for the PVA ASDs with subsequent precipitation and for the HPMCAS ASDs a suppressed dissolution. In order to utilize the unexploited potential of the binary ASDs, the PVA ASDs were combined with HPMCAS either predissolved or added as powder and also formulated as ternary ASD. We successfully generated a solid formulation consisting of the powdered PVA ASD and HPMCAS powder, which was superior in monophasic non-sink dissolution and biorelevant biphasic dissolution studies compared to the binary and ternary ASDs.
Collapse
|
36
|
Sinko PD, Harris S, Salehi N, Meyer PJ, Amidon GL, Amidon GE. Ultrathin, Large-Area Membrane Diffusion Cell for pH-Dependent Simultaneous Dissolution and Absorption Studies. Mol Pharm 2020; 17:2319-2328. [DOI: 10.1021/acs.molpharmaceut.0c00040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Patrick D. Sinko
- Department of Pharmacy, Uppsala Biomedical Centre, Uppsala University, P.O. Box 580, SE-751 23 Uppsala, Sweden
| | | | | | | | | | | |
Collapse
|
37
|
Hate SS, Reutzel-Edens SM, Taylor LS. Absorptive Dissolution Testing: An Improved Approach to Study the Impact of Residual Crystallinity on the Performance of Amorphous Formulations. J Pharm Sci 2020; 109:1312-1323. [DOI: 10.1016/j.xphs.2019.11.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/24/2019] [Accepted: 11/15/2019] [Indexed: 12/25/2022]
|
38
|
Al-Obaidi H, Kowalczyk RM, Kalgudi R, Zariwala MG. Griseofulvin solvate solid dispersions with synergistic effect against fungal biofilms. Colloids Surf B Biointerfaces 2019; 184:110540. [DOI: 10.1016/j.colsurfb.2019.110540] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/27/2019] [Accepted: 09/29/2019] [Indexed: 01/22/2023]
|
39
|
Elkhabaz A, Moseson DE, Brouwers J, Augustijns P, Taylor LS. Interplay of Supersaturation and Solubilization: Lack of Correlation between Concentration-Based Supersaturation Measurements and Membrane Transport Rates in Simulated and Aspirated Human Fluids. Mol Pharm 2019; 16:5042-5053. [PMID: 31638397 DOI: 10.1021/acs.molpharmaceut.9b00956] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Supersaturating formulations are increasingly being used to improve the absorption of orally administered poorly water-soluble drugs. To better predict outcomes in vivo, we must be able to accurately determine the degree of supersaturation in complex media designed to provide a surrogate for the gastrointestinal environment. Herein, we demonstrate that relying on measurements based on consideration of the total dissolved concentration leads to underestimation of supersaturation and consequently membrane transport rates. Crystalline and amorphous solubilities of two compounds, atazanavir and posaconazole, were evaluated in six different media. Concurrently, diffusive flux measurements were performed in a side-by-side diffusion cell to determine the activity-based supersaturation by evaluating membrane transport rates at the crystalline and amorphous solubilities. Solubility values were found to vary in each medium because of different solubilization capacities. Concentration-based supersaturation ratios were also found to vary for the different media. Activity-based measurements, however, were largely independent of the medium, leading to relatively constant values for the estimated supersaturation. These findings have important consequences for modeling and prediction of supersaturation impact on the absorption rate as well as for better defining the thermodynamic driving force for crystallization in complex media.
Collapse
Affiliation(s)
- Ahmed Elkhabaz
- Department of Industrial and Physical Pharmacy, College of Pharmacy , Purdue University , West Lafayette , Indiana 47907 , United States
| | - Dana E Moseson
- Department of Industrial and Physical Pharmacy, College of Pharmacy , Purdue University , West Lafayette , Indiana 47907 , United States
| | - Joachim Brouwers
- Drug Delivery and Disposition , KU Leuven , Leuven 3000 , Belgium
| | | | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy , Purdue University , West Lafayette , Indiana 47907 , United States
| |
Collapse
|
40
|
Que C, Lou X, Zemlyanov DY, Mo H, Indulkar AS, Gao Y, Zhang GGZ, Taylor LS. Insights into the Dissolution Behavior of Ledipasvir–Copovidone Amorphous Solid Dispersions: Role of Drug Loading and Intermolecular Interactions. Mol Pharm 2019; 16:5054-5067. [DOI: 10.1021/acs.molpharmaceut.9b01025] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Chailu Que
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Xiaochun Lou
- Drug Product Development, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Dmitry Y. Zemlyanov
- Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana 47907, United States
| | - Huaping Mo
- Purdue Interdepartmental NMR Facility, Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907, United States
| | - Anura S. Indulkar
- Drug Product Development, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Yi Gao
- Science and Technology, Operations, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Geoff G. Z. Zhang
- Drug Product Development, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Lynne S. Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
41
|
Ricarte RG, Van Zee NJ, Li Z, Johnson LM, Lodge TP, Hillmyer MA. Recent Advances in Understanding the Micro- and Nanoscale Phenomena of Amorphous Solid Dispersions. Mol Pharm 2019; 16:4089-4103. [PMID: 31487183 DOI: 10.1021/acs.molpharmaceut.9b00601] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Many pharmaceutical drugs in the marketplace and discovery pipeline suffer from poor aqueous solubility, thereby limiting their effectiveness for oral delivery. The use of an amorphous solid dispersion (ASD), a mixture of an active pharmaceutical ingredient and a polymer excipient, greatly enhances the aqueous dissolution performance of a drug without the need for chemical modification. Although this method is versatile and scalable, deficient understanding of the interactions between drugs and polymers inhibits ASD rational design. This current Review details recent progress in understanding the mechanisms that control ASD performance. In the solid-state, the use of high-resolution theoretical, computational, and experimental tools resolved the influence of drug/polymer phase behavior and dynamics on stability during storage. During dissolution in aqueous media, novel characterization methods revealed that ASDs can form complex nanostructures, which maintain and improve supersaturation of the drug. The studies discussed here illustrate that nanoscale phenomena, which have been directly observed and quantified, strongly affect the stability and bioavailability of ASD systems, and provide a promising direction for optimizing drug/polymer formulations.
Collapse
Affiliation(s)
- Ralm G Ricarte
- Molecular, Macromolecular Chemistry, and Materials Laboratory, CNRS, ESPCI-Paris , PSL Research University , 10 Rue Vauquelin , 75005 Paris , France
| | | | | | | | | | | |
Collapse
|
42
|
Successful oral delivery of poorly water-soluble drugs both depends on the intraluminal behavior of drugs and of appropriate advanced drug delivery systems. Eur J Pharm Sci 2019; 137:104967. [PMID: 31252052 DOI: 10.1016/j.ejps.2019.104967] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/27/2019] [Accepted: 06/21/2019] [Indexed: 12/11/2022]
Abstract
Poorly water-soluble drugs continue to be a problematic, yet important class of pharmaceutical compounds for treatment of a wide range of diseases. Their prevalence in discovery is still high, and their development is usually limited by our lack of a complete understanding of how the complex chemical, physiological and biochemical processes that occur between administration and absorption individually and together impact on bioavailability. This review defines the challenge presented by these drugs, outlines contemporary strategies to solve this challenge, and consequent in silico and in vitro evaluation of the delivery technologies for poorly water-soluble drugs. The next steps and unmet needs are proposed to present a roadmap for future studies for the field to consider enabling progress in delivery of poorly water-soluble compounds.
Collapse
|