1
|
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
|
2
|
Ueda K, Moseson DE, Taylor LS. Amorphous solubility advantage: Theoretical considerations, experimental methods, and contemporary relevance. J Pharm Sci 2025; 114:18-39. [PMID: 39222748 DOI: 10.1016/j.xphs.2024.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Twenty-five years ago, Hancock and Parks asked a provocative question: "what is the true solubility advantage for amorphous pharmaceuticals?" Difficulties in determining the amorphous solubility have since been overcome due to significant advances in theoretical understanding and experimental methods. The amorphous solubility is now understood to be the concentration after the drug undergoes liquid-liquid or liquid-glass phase separation, forming a water-saturated drug-rich phase in metastable equilibrium with an aqueous phase containing molecularly dissolved drug. While crystalline solubility is an essential parameter impacting the absorption of crystalline drug formulations, amorphous solubility is a vital factor for considering absorption from supersaturating formulations. However, the amorphous solubility of drugs is complex, especially in the presence of formulation additives and gastrointestinal components, and concentration-based measurements may not indicate the maximum drug thermodynamic activity. This review discusses the concept of the amorphous solubility advantage, including a historical perspective, theoretical considerations, experimental methods for amorphous solubility measurement, and the contribution of supersaturation and amorphous solubility to drug absorption. Leveraging amorphous solubility and understanding the associated physicochemical principles can lead to more effective development strategies for poorly water-soluble drugs, ultimately benefiting therapeutic outcomes.
Collapse
Affiliation(s)
- Keisuke Ueda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Dana E Moseson
- Worldwide Research and Development, Pfizer, Inc., Groton, CT 06340, United States
| | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
3
|
Barr KE, Ohnsorg ML, Liberman L, Corcoran LG, Sarode A, Nagapudi K, Feder CR, Bates FS, Reineke TM. Drug-Polymer Nanodroplet Formation and Morphology Drive Solubility Enhancement of GDC-0810. Bioconjug Chem 2024; 35:499-516. [PMID: 38546823 DOI: 10.1021/acs.bioconjchem.4c00018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2024]
Abstract
Nanodroplet formation is important to achieve supersaturation of active pharmaceutical ingredients (APIs) in an amorphous solid dispersion. The aim of the current study was to explore how polymer composition, architecture, molar mass, and surfactant concentration affect polymer-drug nanodroplet morphology with the breast cancer API, GDC-0810. The impact of nanodroplet size and morphology on dissolution efficacy and drug loading capacity was explored using polarized light microscopy, dynamic light scattering, and cryogenic transmission electron microscopy. Poly(N-isopropylacrylamide-stat-N,N-dimethylacrylamide) (PND) was synthesized as two linear derivatives and two bottlebrush derivatives with carboxylated or PEGylated end-groups. Hydroxypropyl methylcellulose acetate succinate grade MF (HPMCAS-MF) and poly(vinylpyrrolidone-co-vinyl acetate) (PVPVA) were included as commercial polymer controls. We report the first copolymerization synthesis of a PVPVA bottlebrush copolymer, which was the highest performing excipient in this study, maintaining 688 μg/mL GDC-0810 concentration at 60 wt % drug loading. This is likely due to strong polymer-drug noncovalent interactions and the compaction of GDC-0810 along the PVPVA bottlebrush backbone. Overall, it was observed that the most effective formulations had a hydrodynamic radius less than 25 nm with tightly compacted nanodroplet morphologies.
Collapse
Affiliation(s)
- Kaylee E Barr
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Monica L Ohnsorg
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Lucy Liberman
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Louis G Corcoran
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Apoorva Sarode
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, California 94080, United States
| | - Karthik Nagapudi
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, California 94080, United States
| | - Christina R Feder
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, California 94080, United States
| | - Frank S Bates
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
4
|
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
|
5
|
Liu YS, Della Rocca J, Schenck L, Koynov A, Sifri RJ, Winston MS, Frank DS. Poly(vinylpyridine- co-vinylpyridine N-oxide) Excipients Mediate Rapid Dissolution and Sustained Supersaturation of Posaconazole Amorphous Solid Dispersions. Mol Pharm 2024; 21:1182-1191. [PMID: 38323546 DOI: 10.1021/acs.molpharmaceut.3c00789] [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: 02/08/2024]
Abstract
The chemical structure of excipients molecularly mixed in an amorphous solid dispersion (ASD) has a significant impact on properties of the ASD including dissolution behavior, physical stability, and bioavailability. Polymers used in ASDs require a balance between hydrophobic and hydrophilic functionalities to ensure rapid dissolution of the amorphous dispersion as well as sustained supersaturation of the drug in solution. This work demonstrates the use of postpolymerization functionalization of poly(vinylpyridine) excipients to elucidate the impact of polymer properties on the dissolution behavior of amorphous dispersions containing posaconazole. It was found that N-oxidation of pyridine functionalities increased the solubility of poly(vinylpyridine) derivatives in neutral aqueous conditions and allowed for nanoparticle formation which supplied posaconazole into solution at concentrations exceeding those achieved by more conventional excipients such as hydroxypropyl methylcellulose acetate succinate (HPMCAS) or Eudragit E PO. By leveraging these functional modifications of the parent poly(vinylpyridine) excipient to increase polymer hydrophilicity and minimize the effect of polymer on pH, a new polymeric excipient was optimized for rapid dissolution and supersaturation maintenance for a model compound.
Collapse
Affiliation(s)
- Yu-Sheng Liu
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Joseph Della Rocca
- Oral Formulation Sciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Luke Schenck
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Athanas Koynov
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Renee J Sifri
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Matthew S Winston
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Derek S Frank
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| |
Collapse
|
6
|
Bapat P, Paul S, Thakral NK, Tseng YC, Taylor LS. Does Media Choice Matter When Evaluating the Performance of Hydroxypropyl Methylcellulose Acetate Succinate-Based Amorphous Solid Dispersions? Mol Pharm 2023; 20:5714-5727. [PMID: 37751517 DOI: 10.1021/acs.molpharmaceut.3c00586] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Hydroxypropyl methylcellulose acetate succinate (HPMCAS) is a weakly acidic polymer that is widely used in the formulation of amorphous solid dispersions (ASDs). While the pH-dependent solubility of HPMCAS is widely recognized, the role of other solution properties, including buffer capacity, is less well understood in the context of ASD dissolution. The goal of this study was to elucidate the rate-limiting steps for drug and HPMCAS release from ASDs formulated with two poorly water soluble model drugs, indomethacin and indomethacin methyl ester. The surface area normalized release rate of the drug and/or polymer in a variety of media was determined. The HPMCAS gel layer apparent pH was determined by incorporating pH sensitive dyes into the polymer matrix. Water uptake extent and rate into the ASDs were measured gravimetrically. For neat HPMCAS, the rate-limiting step for polymer dissolution was observed to be the polymer solubility at the polymer-solution interface. This, in turn, was impacted by the gel layer pH which was found to be substantially lower than the bulk solution pH, varying with medium buffer capacity. For the ASDs, the HPMCAS release rate was found to control the drug release rate. However, both drugs reduced the polymer release rate with indomethacin methyl ester having a larger impact. In low buffer capacity media, the presence of the drug had less impact on release rates when compared to observations in higher strength buffers, suggesting changes in the rate-limiting steps for HPMCAS dissolution. The observations made in this study can contribute to the fundamental understanding of acidic polymer dissolution in the presence and absence of a molecularly dispersed lipophilic drug and will help aid in the design of more in vivo relevant release testing experiments.
Collapse
Affiliation(s)
- Pradnya Bapat
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Shubhajit Paul
- Material and Analytical Sciences, Research and Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Naveen K Thakral
- Material and Analytical Sciences, Research and Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Yin-Chao Tseng
- Material and Analytical Sciences, Research and Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
7
|
Deac A, Qi Q, Indulkar AS, Gao Y, Zhang GGZ, Taylor LS. Dissolution Mechanisms of Amorphous Solid Dispersions: A Close Look at the Dissolution Interface. Mol Pharm 2023; 20:2217-2234. [PMID: 36926898 DOI: 10.1021/acs.molpharmaceut.3c00020] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Despite the recent success of amorphous solid dispersions (ASDs) at enabling the delivery of poorly soluble small molecule drugs, ASD-based dosage forms are limited by low drug loading. This is partially due to a sharp decline in drug release from the ASD at drug loadings surpassing the 'limit of congruency' (LoC). In some cases, the LoC is as low as 5% drug loading, significantly increasing the risk of pill burden. Despite efforts to understand the mechanism responsible for the LoC, a clear picture of the molecular processes occurring at the ASD/solution interface remains elusive. In this study, the ASD/solution interface was studied for two model compounds formulated as ASDs with copovidone. The evolution of a gel layer and its phase behavior was captured in situ with fluorescence confocal microscopy, where fluorescent probes were added to label the hydrophobic and hydrophilic phases. Phase separation was detected in the gel layer for most of the ASDs. The morphology of the hydrophobic phase was found to correlate with the release behavior, where a discrete phase resulted in good release and a continuous phase formed a barrier leading to poor release. The continuous phase formed at a lower drug loading for the system with stronger drug-polymer interactions. This was due to incorporation of the polymer into the hydrophobic phase. The study highlights the complex molecular and phase behavior at the ASD/solution interface of copovidone-based ASDs and provides a thermodynamic argument for qualitatively predicting the release behavior based on drug-polymer interactions.
Collapse
Affiliation(s)
- Alexandru Deac
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Qingqing Qi
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Anura S Indulkar
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Yi Gao
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Geoff G Z Zhang
- Development Sciences, 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
|
8
|
Ueda K, Higashi K, Moribe K. Quantitative Analysis of Drug Supersaturation Region by Temperature-Variable Nuclear Magnetic Resonance Measurements, Part 2: Effects of Solubilizer. Mol Pharm 2023; 20:1872-1883. [PMID: 36939568 DOI: 10.1021/acs.molpharmaceut.3c00050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
This study utilized temperature-variable nuclear magnetic resonance (NMR) spectroscopy to investigate the effects of a solubilizing agent on the ketoprofen (KTP) supersaturation region. Quantitative NMR analysis showed that the solubilizing agent cetyltrimethylammonium bromide (CTAB) increased both the crystalline and amorphous solubilities of KTP, shifting the KTP supersaturation region to a higher KTP concentration range. The amorphous solubility of KTP was found to be independent of the enantiomeric composition of KTP, even in the presence of CTAB. However, the supersaturation region of the S-enantiomer of KTP (s-KTP) in CTAB solutions was smaller than that of the racemic form of KTP (rac-KTP), likely because of the higher crystalline solubility of s-KTP. When KTP formed a KTP-rich phase via liquid-liquid phase separation from KTP-supersaturated solutions, CTAB was observed to be distributed into the KTP-rich phase, decreasing the chemical potential of KTP and the maximum thermodynamic activity of KTP in the aqueous phase. Additionally, the incorporation of CTAB into the KTP-rich phase diminished the solubilization effect of CTAB micelles in the aqueous phase, narrowing the KTP supersaturation region to a greater extent at higher KTP dose concentrations. Furthermore, the upper-temperature limit of the supersaturated dissolvable region of KTP was lowered in the presence of CTAB, which was rationalized by the melting point depression of the KTP crystal upon mixing with CTAB. The findings of this study highlight the importance of considering the molecular-level impact of solubilizing agents on the drug supersaturation region to fully exploit the potential benefits of supersaturated formulations.
Collapse
Affiliation(s)
- Keisuke Ueda
- 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
|
9
|
Ueda K, Higashi K, Moribe K. Quantitative Analysis of Drug Supersaturation Region by Temperature-Variable Nuclear Magnetic Resonance Measurements, Part 1: Effects of Polymer and Drug Chiralities. Mol Pharm 2023; 20:1861-1871. [PMID: 36939575 DOI: 10.1021/acs.molpharmaceut.2c00924] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
We examined the effects of the polymer-additive and drug chiralities on the ketoprofen (KTP) supersaturation region using temperature-variable nuclear magnetic resonance (NMR). Quantitative NMR analysis revealed that the racemic KTP and corresponding S-enantiomer (rac- and s-KTP) exhibited similar amorphous solubilities in a buffer, while the crystalline solubility of s-KTP was higher than that of rac-KTP. Therefore, rac-KTP exhibited a larger supersaturation region than s-KTP. In contrast, polyvinylpyrrolidone (PVP) reduced the amorphous solubility of both rac- and s-KTP, whereas the crystalline solubility of KTP remained unchanged. Partitioning PVP into the KTP-rich phase reduced the chemical potential of KTP in the KTP-rich phase and the amorphous solubility of KTP. At higher temperatures, the distribution of PVP into the KTP-rich phase became more significant, which considerably reduced the amorphous solubility. Because the upper limit of the KTP supersaturation decreased, PVP narrowed the KTP supersaturation region. The maximum KTP supersaturation ratio decreased with increasing temperature, and the supersaturated dissolvable area of KTP finally disappeared. The maximum temperature at which KTP can form the supersaturation was lowered by replacing rac- with s-KTP and the addition of PVP. The maximum supersaturation temperature was dominated by the melting behavior of crystalline KTP in an aqueous solution. The present study highlighted that a quantitative understanding of the supersaturation region is essential to determine whether supersaturated formulations are beneficial for improving the oral absorption of poorly water-soluble drugs.
Collapse
Affiliation(s)
- Keisuke Ueda
- 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
|
10
|
Andrews GP, Qian K, Jacobs E, Jones DS, Tian Y. High drug loading nanosized amorphous solid dispersion (NASD) with enhanced in vitro solubility and permeability: Benchmarking conventional ASD. Int J Pharm 2023; 632:122551. [PMID: 36581107 DOI: 10.1016/j.ijpharm.2022.122551] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/06/2022] [Accepted: 12/23/2022] [Indexed: 12/27/2022]
Abstract
Through liquid-liquid phase separation (LLPS), it is possible to generate drug-rich nanoparticles during the dissolution of conventional amorphous solid dispersions (ASDs). These self-generated nanoparticles may improve the oral absorption of poorly water-soluble drugs by enhancing the drug's apparent solubility and effective membrane permeability. However, due to the high concentration threshold required for LLPS, conventional ASDs that can consistently generate drug-rich nanoparticles during dissolution are rare. More importantly, the quality of these meta-stable drug-rich nanoparticles is hard to control during dissolution, leading to inconsistency in formulation performances. This work has described a continuous twin-screw extrusion process capable of producing nanosized ASD (NASD) formulations that can offer better solubility and permeability enhancements over conventional ASD formulations. Two polymeric carriers, polyvinylpyrrolidone-co-vinyl acetate (PVPVA) and hydroxypropyl methylcellulose acetate succinate (HPMCAS), with a model hydrophobic drug celecoxib (BCS II), were formulated into both ASD and NASD formulations. Compared to the conventional ASD formulation, the prefabricated NASD (sizes ranging between 40 and 200 nm) embedded within a polyol matrix can be rapidly dispersed into a nanoparticle suspension in the presence of aqueous media. The resulting NASDs achieved drug loadings up to 80 % w/w and a maximum of 98 % encapsulation efficiency. Because of the TSE platform's high drug-loading capacity and high scalability, the developed method may be useful for continuously producing personalized nanomedicines.
Collapse
Affiliation(s)
- Gavin P Andrews
- School of Pharmacy, Queen's University Belfast, BT9 7BL, United Kingdom
| | - Kaijie Qian
- School of Pharmacy, Queen's University Belfast, BT9 7BL, United Kingdom
| | - Esther Jacobs
- School of Pharmacy, Queen's University Belfast, BT9 7BL, United Kingdom
| | - David S Jones
- School of Pharmacy, Queen's University Belfast, BT9 7BL, United Kingdom
| | - Yiwei Tian
- School of Pharmacy, Queen's University Belfast, BT9 7BL, United Kingdom.
| |
Collapse
|
11
|
Yang R, Zhang GGZ, Zemlyanov DY, Purohit HS, Taylor LS. Release Mechanisms of Amorphous Solid Dispersions: Role of Drug-Polymer Phase Separation and Morphology. J Pharm Sci 2023; 112:304-317. [PMID: 36306863 DOI: 10.1016/j.xphs.2022.10.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/07/2022] [Accepted: 10/20/2022] [Indexed: 11/13/2022]
Abstract
Formulating poorly soluble molecules as amorphous solid dispersions (ASDs) is an effective strategy to improve drug release. However, drug release rate and extent tend to rapidly diminish with increasing drug loading (DL). The poor release at high DLs has been postulated to be linked to the process of amorphous-amorphous phase separation (AAPS), although the exact connection between phase separation and release properties remains somewhat unclear. Herein, release profiles of ASDs formulated with ritonavir (RTV) and polyvinylpyrrolidone/vinyl acetate (PVPVA) at different DLs were determined using surface normalized dissolution. Surface morphologies of partially dissolved ASD compacts were evaluated with confocal fluorescence microscopy, using Nile red and Alexa Fluor 488 as fluorescence markers to track the hydrophobic and hydrophilic phases respectively. ASD phase behavior during hydration and release of components were also visualized in real time using a newly developed in situ confocal fluorescence microscopy method. RTV-PVPVA ASDs showed complete and rapid drug release below 30% DL, partial drug release at 30% DL and no drug release above 30% DL. It was observed that formation of discrete drug-rich droplets at lower DLs led to rapid and congruent release of both drug and polymer, whereas formation of continuous drug-rich phase at the ASD matrix-solution interface was the cause of poor release above certain DLs. Thus, the domain size and interconnectivity of phase separated drug-rich domains appear to be critical factors impacting drug release from RTV-PVPVPA ASDs.
Collapse
Affiliation(s)
- Ruochen Yang
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
| | - Geoff G Z Zhang
- Drug Product Development, AbbVie Inc., North Chicago, IL, 60064, USA
| | - Dmitry Y Zemlyanov
- Birck Nanotechnology Center, Purdue University, West Lafayette, IN, 47907, USA
| | - Hitesh S Purohit
- Drug Product Development, AbbVie Inc., North Chicago, IL, 60064, USA.
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
12
|
Zhao P, Han W, Shu Y, Li M, Sun Y, Sui X, Liu B, Tian B, Liu Y, Fu Q. Liquid-liquid phase separation drug aggregate: Merit for oral delivery of amorphous solid dispersions. J Control Release 2023; 353:42-50. [PMID: 36414193 DOI: 10.1016/j.jconrel.2022.11.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022]
Abstract
As a promising strategy, amorphous solid dispersion has been extensively employed in improving the oral bioavailability of insoluble drugs. Despite the numerous advantages, the problems associated with supersaturation stability limit its further application. Recently, the formation and stability of the liquid-liquid phase separation drug aggregate (LLPS-DA) have been found to be vital for supersaturation maintenance. An in-depth review of LLPS-DA was required to further explore the supersaturation maintenance mechanism in vivo. Hence, this study aimed to present a short review to introduce the LLPS-DA, highlight the in vivo advantages for oral administration, and discuss the prospects to help understand the in vivo behavior of LLPS-DA.
Collapse
Affiliation(s)
- Peixu Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Wen Han
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Yecheng Shu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Mo Li
- Liaoning Institute for Drug Control, No. 7 Chongshan West Road, Shenyang 110016, China
| | - Yichi Sun
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Xiaofan Sui
- Liaoning Institute for Drug Control, No. 7 Chongshan West Road, Shenyang 110016, China
| | - Bingyang Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Baocheng Tian
- School of Pharmacy, Binzhou Medical University, No. 346, Guanhai Road, Yantai 264003, China
| | - Yanhua Liu
- Department of Pharmaceutics, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
| | - Qiang Fu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
13
|
Fukiage M, Suzuki K, Matsuda M, Nishida Y, Oikawa M, Fujita T, Kawakami K. Inhibition of Liquid-Liquid Phase Separation for Breaking the Solubility Barrier of Amorphous Solid Dispersions to Improve Oral Absorption of Naftopidil. Pharmaceutics 2022; 14:pharmaceutics14122664. [PMID: 36559158 PMCID: PMC9782492 DOI: 10.3390/pharmaceutics14122664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/25/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022] Open
Abstract
Amorphous solid dispersion (ASD) is one of the most promising technologies for improving the oral absorption of poorly soluble compounds. In this study, naftopidil (NFT) ASDs were prepared using vinylpyrrolidone-vinyl acetate copolymer (PVPVA), hydroxypropyl methylcellulose acetate succinate (HPMCAS), and poly(methacrylic acid-co-methyl methacrylate) L100-55 (Eudragit) to improve the dissolution and oral absorption behaviors of NFT. During the dissolution process of ASD, liquid-liquid phase separation (LLPS) may occur when certain requirements are met for providing a maximum quasi-stable concentration achievable by amorphization. The occurrence of LLPS was confirmed in the presence of PVPVA and HPMCAS; however, Eudragit inhibited LLPS owing to its molecular interaction with NFT. Although the dissolution behavior of the Eudragit ASD was found to be markedly poorer than that of other ASDs, it offered the best oral absorption in rats. The findings of the current study highlight the possibility for improving the oral absorption of poorly soluble drugs by this ASD, which should be eliminated from candidate formulations based on the conventional in vitro tests.
Collapse
Affiliation(s)
- Masafumi Fukiage
- Pharmaceutical R&D, Ono Pharmaceutical Co., Ltd., 3-3-1, Sakurai, Shimamoto-cho, Mishima-gun, Osaka 618-8585, Osaka, Japan
- Correspondence: (M.F.); (K.K.); Tel.: +81-75-961-1151 (M.F.); Tel.: +81-29-860-4424 (K.K.)
| | - Kyosuke Suzuki
- Pharmaceutical and ADMET Research Department, Daiichi Sankyo RD Novare Co., Ltd., 1-16-13, Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Maki Matsuda
- Research & Development Division, Towa Pharmaceutical Co., Ltd., 134, Chudoji Minami-machi, Shimogyo-ku, Kyoto 600-8813, Kyoto, Japan
| | - Yohei Nishida
- Technology Research & Development, Sumitomo Pharma Co., Ltd., 33-94, Enoki-cho, Suita, Osaka 564-0053, Osaka, Japan
| | - Michinori Oikawa
- Pharmaceutical Development Department, Sawai Pharmaceutical Co., Ltd., 5-2-30, Miyahara, Yodogawa-ku, Osaka 532-0003, Osaka, Japan
| | - Takuya Fujita
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Kyoto 525-8577, Shiga, Japan
| | - Kohsaku Kawakami
- Research Center for Functionals Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba 305-0044, Ibaraki, Japan
- Correspondence: (M.F.); (K.K.); Tel.: +81-75-961-1151 (M.F.); Tel.: +81-29-860-4424 (K.K.)
| |
Collapse
|
14
|
Designing synergistic crystallization inhibitors: Bile salt derivatives of cellulose with enhanced hydrophilicity. Carbohydr Polym 2022; 292:119680. [PMID: 35725174 DOI: 10.1016/j.carbpol.2022.119680] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 05/10/2022] [Accepted: 05/28/2022] [Indexed: 11/23/2022]
Abstract
Crystallization inhibitors in amorphous solid dispersions (ASD) enable metastable supersaturated drug solutions that persist for a physiologically relevant time. Olefin cross-metathesis (CM) has successfully provided multifunctional cellulose-based derivatives as candidate ASD matrix polymers. In proof of concept studies, we prepared hydrophobic bile salt/cellulose adducts by CM with naturally occurring bile salts. We hypothesized that increased hydrophilicity would enhance the ability of these conjugates to maximize bioactive supersaturation. Their selective preparation presents a significant synthetic challenge, given polysaccharide reactivity and polysaccharide and bile salt complexity. We prepared such derivatives using a more hydrophilic hydroxypropyl cellulose (HPC) backbone, employing a pent-4-enyl tether (Pen) for appending bile acids. We probed structure-property relationships by varying the nature and degree of substitution of the bile acid substituent (lithocholic or deoxycholic acid). These conjugates are indeed synergistic inhibitors, as demonstrated with the fast-crystallizing prostate cancer drug, enzalutamide. The lithocholic acid methyl ester derivative, AcrMLC-PenHHPCPen (0.64), increased induction time 68 fold vs. drug alone.
Collapse
|
15
|
Nambiar AG, Singh M, Mali AR, Serrano DR, Kumar R, Healy AM, Agrawal AK, Kumar D. Continuous Manufacturing and Molecular Modeling of Pharmaceutical Amorphous Solid Dispersions. AAPS PharmSciTech 2022; 23:249. [PMID: 36056225 DOI: 10.1208/s12249-022-02408-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 08/24/2022] [Indexed: 11/30/2022] Open
Abstract
Amorphous solid dispersions enhance solubility and oral bioavailability of poorly water-soluble drugs. The escalating number of drugs with poor aqueous solubility, poor dissolution, and poor oral bioavailability is an unresolved problem that requires adequate interventions. This review article highlights recent solubility and bioavailability enhancement advances using amorphous solid dispersions (ASDs). The review also highlights the mechanism of enhanced dissolution and the challenges faced by ASD-based products, such as stability and scale-up. The role of process analytical technology (PAT) supporting continuous manufacturing is highlighted. Accurately predicting interactions between the drug and polymeric carrier requires long experimental screening methods, and this is a space where computational tools hold significant potential. Recent advancements in data science, computational tools, and easy access to high-end computation power are set to accelerate ASD-based research. Hence, particular emphasis has been given to molecular modeling techniques that can address some of the unsolved questions related to ASDs. With the advancement in PAT tools and artificial intelligence, there is an increasing interest in the continuous manufacturing of pharmaceuticals. ASDs are a suitable option for continuous manufacturing, as production of a drug product from an ASD by direct compression is a reality, where the addition of multiple excipients is easy to avoid. Significant attention is necessary for ongoing clinical studies based on ASDs, which is paving the way for the approval of many new ASDs and their introduction into the market.
Collapse
Affiliation(s)
- Amritha G Nambiar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Maan Singh
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Abhishek R Mali
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | | | - Rajnish Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Anne Marie Healy
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - Ashish Kumar Agrawal
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Dinesh Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India.
| |
Collapse
|
16
|
Supramolecular encapsulation of nanocrystalline Schiff bases into β-cyclodextrin for multifold enrichment of bio-potency. Carbohydr Polym 2022; 291:119614. [DOI: 10.1016/j.carbpol.2022.119614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/13/2022] [Accepted: 05/10/2022] [Indexed: 01/14/2023]
|
17
|
Ueda K, Higashi K, Moribe K. Unusual Correlation between the Apparent Amorphous Solubility of a Drug and Solubilizer Concentration Revealed by NMR Analysis. Mol Pharm 2022; 19:3336-3349. [PMID: 35924819 DOI: 10.1021/acs.molpharmaceut.2c00478] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Herein, we investigated the effect of the solubilizers, cetyltrimethylammonium bromide (CTAB) and amino methacrylate copolymer (Eudragit E PO, EUD-E), on the apparent amorphous solubility of ketoprofen (KTP) and free KTP concentrations in an aqueous phase when a KTP-rich phase was generated by liquid-liquid phase separation. Quantitative analysis by solution nuclear magnetic resonance (NMR) revealed that the apparent amorphous solubility of KTP increased with increasing EUD-E concentrations by the solubilization of KTP into the EUD-E micelles; this was reminiscent of the improvement in the apparent crystalline solubility of KTP observed when EUD-E was added. In contrast, the apparent amorphous solubility of KTP decreased with increasing CTAB concentrations, although the solubilizing ability of CTAB was stronger than that of EUD-E when the KTP-rich phase was absent. NMR analysis revealed that CTAB was distributed into the KTP-rich phase to a relatively large extent. This resulted in a significant reduction of the chemical potential of KTP in the KTP-rich phase in the CTAB solution. Thus, the maximum free KTP concentration in the aqueous phase was reduced more significantly in the CTAB solution than in the EUD-E solution. Moreover, the solubilization effect of KTP by the CTAB micelles in the aqueous phase was drastically diminished due to the distribution of CTAB into the KTP-rich phase. As a result, the apparent amorphous solubility of KTP reached a minimum at a CTAB concentration of 200 μg/mL. A further increase in the CTAB concentration resulted in an improvement in the apparent amorphous solubility of KTP due to the solubilization effect of CTAB remaining in the aqueous phase. The present study highlights the impact of solubilizer selection on the apparent amorphous solubility and attainable supersaturation of the drug, which should be considered during the development of supersaturating formulations to obtain preferable oral absorption.
Collapse
Affiliation(s)
- Keisuke Ueda
- 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
|
18
|
Yang R, Zhang GGZ, Kjoller K, Dillon E, Purohit HS, Taylor LS. Phase separation in surfactant-containing amorphous solid dispersions: Orthogonal analytical methods to probe the effects of surfactants on morphology and phase composition. Int J Pharm 2022; 619:121708. [PMID: 35364219 DOI: 10.1016/j.ijpharm.2022.121708] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/23/2022] [Accepted: 03/26/2022] [Indexed: 11/19/2022]
Abstract
Amorphous-amorphous phase separation (AAPS) is an important phase transition process for amorphous solid dispersion (ASD) performance both in terms of drug release as well as physical and chemical stability during storage. Addition of surfactants to ASD systems can impact both of these processes. One possible mechanism through which surfactants affect ASD performance is via their impact on AAPS. Unfortunately, despite their increasing usage in ASD formulations, the effect of surfactant on AAPS is still poorly understood, and there are limited analytical techniques that provide microstructural and composition information about phase separated ASDs. In this study, the impact of four surfactants (sodium dodecyl sulfate, Tween 80, Span 20 and Span 85) on water-induced phase separation in ASDs formulated with ritonavir and polyvinylpyrrolidone/vinyl acetate (PVPVA) was investigated using a variety of orthogonal analytical methods. Transparent films of ASDs with different compositions were prepared by spin coating. Fluorescence confocal microscopy in combination with an in situ humidity chamber was used to monitor the kinetics and morphology of phase separation following exposure to high relative humidity. Optical photothermal IR analysis of phase separated films enabled characterization of domain composition and surfactant distribution. Liquid-liquid phase separation concentration, zeta potential and solution nuclear magnetic resonance spectroscopy measurements enabled interpretation of interaction with and partition of surfactants into the drug-rich phase. It was found that phase separation kinetics and morphology were notably changed by the surfactants. Further, the surfactants showed different affinities for the drug-rich versus the aqueous/polymer-rich phases. The employed analytical techniques were found to be complementary in providing insight into surfactant location in phase separated systems. This study highlights the complexity of phase separation, especially in the presence of surfactants, and provides a foundation to understand the impact of AAPS on ASD performance.
Collapse
Affiliation(s)
- Ruochen Yang
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Geoff G Z Zhang
- Drug Product Development, AbbVie Inc., North Chicago, IL 60064, USA
| | - Kevin Kjoller
- Photothermal Spectroscopy Corp, Santa Barbara, CA 93101, USA
| | - Eoghan Dillon
- Photothermal Spectroscopy Corp, Santa Barbara, CA 93101, USA
| | - Hitesh S Purohit
- Drug Product Development, AbbVie Inc., North Chicago, IL 60064, USA.
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
19
|
Impact of Surfactants on the Performance of Clopidogrel-Copovidone Amorphous Solid Dispersions: Increased Drug Loading and Stabilization of Nanodroplets. Pharm Res 2022; 39:167-188. [PMID: 35013849 DOI: 10.1007/s11095-021-03159-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/17/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE Surfactants are increasingly being added to amorphous solid dispersion (ASDs) formulations to enhance processability and release performance. The goal of the current work was to investigate the impact of cationic, anionic and non-ionic surfactants on the rate and extent of clopidogrel (CPD) release from copovidone-based ASDs. METHODS CPD release was evaluated for ASDs with different drug loadings using a surface normalized intrinsic dissolution apparatus. Studies were also carried out using dynamic light scattering, zeta potential measurements, and nuclear magnetic resonance spectroscopy to probe the impact of surfactants on drug-rich nanodroplet physical stability and clopidogrel-surfactant interactions. RESULTS CPD ASDs showed good release for drug loadings as high as 40%, before the release fell off a cliff at higher drug loadings. Only sodium dodecyl sulfate, added at a 5% level, was able to improve the release at 50% drug loading, with other surfactants proving to be ineffective. However, some of the surfactants evaluated did show some benefits in improving nanodroplet stability against size enlargement. Ionic and non-ionic surfactants were observed to interact differently with CPD-rich nanodroplets, and variations in the kinetics and morphology of water-induced phase separation were noted in the presence and absence of surfactants in ASD films. CONCLUSIONS In summary, addition of surfactants to ASD formulations may lead to some improvements in formulation performance, but predictive capabilities and mechanisms of surfactant effect still require further studies.
Collapse
|
20
|
Shi Q, Li F, Yeh S, Moinuddin SM, Xin J, Xu J, Chen H, Ling B. Recent Advances in Enhancement of Dissolution and Supersaturation of Poorly Water-Soluble Drug in Amorphous Pharmaceutical Solids: A Review. AAPS PharmSciTech 2021; 23:16. [PMID: 34893936 DOI: 10.1208/s12249-021-02137-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/07/2021] [Indexed: 12/19/2022] Open
Abstract
Amorphization is one of the most effective pharmaceutical approaches to enhance the dissolution and oral bioavailability of poorly water-soluble drugs. In recent years, amorphous formulations have been experiencing rapid development both in theoretical and practical application. Based on using different types of stabilizing agents, amorphous formulations can be mainly classified as polymer-based amorphous solid dispersion, coamorphous formulation, mesoporous silica-based amorphous formulation, etc. This paper summarizes recent advances in the dissolution and supersaturation of these amorphous formulations. Moreover, we also highlight the roles of stabilizing agents such as polymers, low molecular weight co-formers, and mesoporous silica. Maintaining supersaturation in solution is a key factor for the enhancement of dissolution profile and oral bioavailability, and thus, the strategies and challenges for maintaining supersaturation are also discussed. With an in-depth understanding of the inherent mechanisms of dissolution behaviors, the design of amorphous pharmaceutical formulations will become more scientific and reasonable, leading to vigorous development of commercial amorphous drug products.
Collapse
|
21
|
Liu Y, Chen X, Yu DG, Liu H, Liu Y, Liu P. Electrospun PVP-Core/PHBV-Shell Fibers to Eliminate Tailing Off for an Improved Sustained Release of Curcumin. Mol Pharm 2021; 18:4170-4178. [PMID: 34582196 DOI: 10.1021/acs.molpharmaceut.1c00559] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tailing off release in the sustained release of water-insoluble curcumin (Cur) is a significant challenge in the drug delivery system. As a novel solution, core-shell nanodrug containers have aroused many interests due to their potential improvement in drug-sustained release. In this work, a biodegradable polymer, poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV), and hydrophilic polyvinylpyrrolidone (PVP) were exploited as drug delivery carriers by coaxial electrospinning, and the core-shell drug-loaded fibers exhibited improved sustained release of Cur. A cylindrical morphology and a clear core-shell structure were observed by scanning and transmission electron microscopies. The X-ray diffraction pattern and infrared spectroscopy revealed that Cur existed in amorphous form due to its good compatibility with PHBV and PVP. The in vitro drug release curves confirmed that the core-shell container manipulated Cur in a faster drug release process than that in the traditional PHBV monolithic container. The combination of the material and structure forms a novel nanodrug container with a better sustained release of water-insoluble Cur. This strategy is beneficial for exploiting more functional biomedical materials to improve the drug release behavior.
Collapse
Affiliation(s)
- Yubo Liu
- School of Materials Science & Engineering, University of Shanghai for Science & Technology, Shanghai 200093, China
| | - Xiaohong Chen
- School of Materials Science & Engineering, University of Shanghai for Science & Technology, Shanghai 200093, China.,Shanghai Engineering Technology Research Center for High-Performance Medical Device Materials, Shanghai 200093, China
| | - Deng-Guang Yu
- School of Materials Science & Engineering, University of Shanghai for Science & Technology, Shanghai 200093, China.,Shanghai Engineering Technology Research Center for High-Performance Medical Device Materials, Shanghai 200093, China
| | - Hang Liu
- School of Materials Science & Engineering, University of Shanghai for Science & Technology, Shanghai 200093, China
| | - Yuyang Liu
- School of Materials Science & Engineering, University of Shanghai for Science & Technology, Shanghai 200093, China
| | - Ping Liu
- School of Materials Science & Engineering, University of Shanghai for Science & Technology, Shanghai 200093, China.,Shanghai Engineering Technology Research Center for High-Performance Medical Device Materials, Shanghai 200093, China
| |
Collapse
|
22
|
Ueda K, Higashi K, Moribe K, Taylor LS. Variable-Temperature NMR Analysis of the Thermodynamics of Polymer Partitioning between Aqueous and Drug-Rich Phases and Its Significance for Amorphous Formulations. Mol Pharm 2021; 19:100-114. [PMID: 34702040 DOI: 10.1021/acs.molpharmaceut.1c00664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We previously reported that the polymers used in amorphous solid dispersion (ASD) formulations, such as polyvinylpyrrolidone (PVP), polyvinylpyrrolidone/vinyl acetate (PVP-VA), and hypromellose (HPMC), distribute into the drug-rich phase of ibuprofen (IBP) formed by liquid-liquid phase separation, resulting in a reduction in the maximum drug supersaturation in the aqueous phase. Herein, the mechanism underlying the partitioning of the polymer into the drug-rich phase was investigated from a thermodynamic perspective. The dissolved IBP concentration in the aqueous phase and the amount of polymer distributed into the IBP-rich phase were quantitatively analyzed in IBP-supersaturated solutions containing different polymers using variable-temperature solution-state nuclear magnetic resonance (NMR) spectroscopy. The polymer weight ratio in the IBP-rich phase increased at higher temperatures, leading to a more notable reduction of IBP amorphous solubility. Among the polymers, the amorphous solubility reduction was the greatest for the PVP-VA solution at lower temperatures, while HPMC reduced the amorphous solubility to the greatest extent at higher temperatures. The change in the order of polymer impact on the amorphous solubility resulted from the differences in the temperature dependency of polymer partitioning. The van't Hoff plot of the polymer partition coefficient revealed that both enthalpy and entropy changes for polymer transfer into the IBP-rich phase from the aqueous phase (ΔHaqueous→IBP-rich and ΔSaqueous→IBP-rich) gave positive values for most of the measured temperature range, indicating that polymer partitioning into the IBP-rich phase was an endothermic but entropically favorable process. The polymer transfer into the IBP-rich phase was more endothermic for HPMC than for PVP and PVP-VA. The solid-state NMR analysis of the IBP/polymer ASD implied that the newly formed IBP/polymer interactions in the IBP-rich phase upon polymer incorporation were weaker for HPMC, providing a rationale for the larger positive transfer enthalpy for HPMC. The change in Gibbs free energy for polymer transfer (ΔGaqueous→IBP-rich) showed negative values across the experimental temperature range, decreasing with an increase in temperature, indicating that the distribution of the polymer into the IBP-rich phase is favored at higher temperatures. Moreover, ΔGaqueous→IBP-rich for HPMC showed the greatest decrease with the temperature, likely reflecting the temperature-induced dehydration of HPMC in the aqueous phase. This study contributes fundamental insights into the phenomenon of polymer partitioning into drug-rich phases, furthering the understanding of achievable supersaturation levels and ultimately providing information on polymer selection for ASD formulations.
Collapse
Affiliation(s)
- Keisuke Ueda
- 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
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
23
|
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
|
24
|
Ueda K, Higashi K, Moribe K. Amorphous Drug Solubility and Maximum Free Drug Concentrations in Cyclodextrin Solutions: A Quantitative Study Using NMR Diffusometry. Mol Pharm 2021; 18:2764-2776. [PMID: 34180226 DOI: 10.1021/acs.molpharmaceut.1c00311] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cyclodextrin (CD) has been widely used as a solubilizing agent for poorly water-soluble drugs. In the present study, the effect of CD on the amorphous drug solubility and the maximum thermodynamic activity of the drug in the aqueous phase when the drug concentration exceeded the liquid-liquid phase separation (LLPS) concentration was investigated using three chemically diverse CDs, β-cyclodextrin (β-CD), dimethyl-β-CD (DM-β-CD), and hydroxypropyl-β-CD (HP-β-CD). The amorphous solubility of ibuprofen (IBP) increased substantially linearly with the increase in the CD concentration due to IBP/CD complex formation. Surprisingly, although the crystalline solubility of IBP in the β-CD solution reached a plateau at β-CD concentrations above 3 mM (BS-type solubility diagram) because of the limited crystalline solubility of the IBP/β-CD complex, the amorphous solubility of IBP increased linearly even when the β-CD concentration was higher than 3 mM. The amorphous solubility of IBP in CD solutions was influenced primarily by the phase separation of the IBP-supersaturated solution to the aqueous phase and the other phase mainly composed of IBP, namely, the IBP-rich phase, via LLPS. NMR spectroscopy revealed that DM-β-CD was distributed into the IBP-rich phase when the IBP concentration exceeded its amorphous solubility, while β-CD and HP-β-CD showed minimal mixing with the IBP-rich phase. NMR diffusometry showed that the maximum free IBP concentration was reduced in the DM-β-CD solution compared to that in the buffer. The mixing of DM-β-CD with the IBP-rich phase reduced the chemical potential of IBP in the IBP-rich phase, which in turn reduced the maximum thermodynamic activity of IBP in the aqueous phase. In contrast, the maximum free IBP concentration was unchanged when β-CD or HP-β-CD was present. The hydrophobic nature of the DM-β-CD substituent may contribute to its partitioning into the IBP-rich phase. The present study highlights the impact of CD on the maximum thermodynamic activity of drugs as well as the apparent amorphous solubility of the drug. This aspect should be considered for improving the effective absorption of poorly water-soluble drugs.
Collapse
Affiliation(s)
- Keisuke Ueda
- 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
|
25
|
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
|
26
|
Saboo S, Bapat P, Moseson DE, Kestur US, Taylor LS. Exploring the Role of Surfactants in Enhancing Drug Release from Amorphous Solid Dispersions at Higher Drug Loadings. Pharmaceutics 2021; 13:pharmaceutics13050735. [PMID: 34067666 PMCID: PMC8156319 DOI: 10.3390/pharmaceutics13050735] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/04/2021] [Accepted: 05/10/2021] [Indexed: 12/20/2022] Open
Abstract
To reduce the dosage size of amorphous solid dispersion (ASD)-based formulations, it is of interest to devise formulation strategies that allow increased drug loading (DL) without compromising dissolution performance. The aim of this study was to explore how surfactant addition impacts drug release as a function of drug loading from a ternary ASD, using felodipine as a model poorly soluble compound. The addition of 5% TPGS (d-α-tocopheryl polyethylene glycol 1000 succinate, a surfactant) to felodipine-polyvinylpyrrolidone/vinyl acetate ASDs was found to facilitate rapid and congruent (i.e., simultaneous) release of drug and polymer at higher DLs relative to binary ASDs (drug and polymer only). For binary ASDs, good release was observed for DLs up to <20% DL; this increased to 35% DL with surfactant. Microstructure evolution in ASD films following exposure to 100% relative humidity was studied using atomic force microscopy coupled with nanoscale infrared imaging. The formation of discrete, spherical drug-rich domains in the presence of surfactant appeared to be linked to systems showing congruent and rapid release of drug and polymer. In contrast, a contiguous drug-rich phase was formed for systems without surfactant at higher DLs. This study supports the addition of surfactant to ASD formulations as a strategy to increase DL without compromising release. Furthermore, insights into the potential role of surfactant in altering ASD release mechanisms are provided.
Collapse
Affiliation(s)
- Sugandha Saboo
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; (S.S.); (P.B.); (D.E.M.)
- Oral Formulation Sciences and Technology, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Pradnya Bapat
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; (S.S.); (P.B.); (D.E.M.)
| | - Dana E. Moseson
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; (S.S.); (P.B.); (D.E.M.)
| | - Umesh S. Kestur
- Drug Product Development, Bristol-Myers Squibb Company, One Squib Drive, New Brunswick, NJ 08903, USA;
| | - Lynne S. Taylor
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA; (S.S.); (P.B.); (D.E.M.)
- Correspondence: ; Tel.: +1-765-496-6614
| |
Collapse
|
27
|
Yang R, Mann AKP, Van Duong T, Ormes JD, Okoh GA, Hermans A, Taylor LS. Drug Release and Nanodroplet Formation from Amorphous Solid Dispersions: Insight into the Roles of Drug Physicochemical Properties and Polymer Selection. Mol Pharm 2021; 18:2066-2081. [PMID: 33784104 DOI: 10.1021/acs.molpharmaceut.1c00055] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Dissolution of amorphous solid dispersions (ASD) can lead to the formation of amorphous drug-rich nano species (nanodroplets) via liquid-liquid phase separation or glass-liquid phase separation when the drug concentration exceeds the amorphous solubility. These nanodroplets have been shown to be beneficial for ASD performance both in vitro and in vivo. Thus, understanding the generation and stability of nanodroplets from ASD formulations is important. In this study, the impacts of polymer selection and active pharmaceutical ingredient (API) physicochemical properties (wet glass transition temperature (Tg) and log P) on nanodroplet release were studied. Six APIs with different physicochemical properties were formulated as ASDs with two polymers, polyvinylpyrrolidone/vinyl acetate (PVPVA) and hydroxypropyl methylcellulose acetate succinate (HPMCAS). Their release performance was evaluated using both powder and surface normalized dissolution of compacts. In general, HPMCAS-based dispersions resulted in higher drug release compared to PVPVA-based dispersions. The two polymers also exhibited different trends in nanodroplet formation as a function of drug loading (DL). PVPVA ASDs exhibited a "falling-off-the-cliff" effect, with a dramatic decline in release performance with a small increase in drug loading, while HPMCAS ASDs exhibited a negative "slope" in the release rate as a function of drug loading. For both polymers, low Tg compounds achieved higher levels of nanodroplet formation compared to high Tg compounds. The nanodroplets generated from ASD dissolution were also monitored with dynamic light scattering, and HPMCAS was found to be more effective at stabilizing nanodroplets against size increase. Insights from this study may be used to guide formulation design and selection of excipients based on API physicochemical properties.
Collapse
Affiliation(s)
- Ruochen Yang
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Amanda K P Mann
- Merck & Co., Inc. 2000, Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Tu Van Duong
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - James D Ormes
- Merck & Co., Inc. 2000, Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Grace A Okoh
- Merck & Co., Inc. 2000, Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Andre Hermans
- Merck & Co., Inc. 2000, Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|
28
|
Ueda K, Moseson DE, Pathak V, Taylor LS. Effect of Polymer Species on Maximum Aqueous Phase Supersaturation Revealed by Quantitative Nuclear Magnetic Resonance Spectroscopy. Mol Pharm 2021; 18:1344-1355. [PMID: 33595322 DOI: 10.1021/acs.molpharmaceut.0c01174] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The polymer used in an amorphous solid dispersion (ASD) formulation impacts the maximum achievable drug supersaturation. Herein, the effect of dissolved polymer on drug concentration in the aqueous phase when a drug-rich phase was generated by liquid-liquid phase separation (LLPS) was investigated for different polymers at various concentrations of drug and polymer. Solution nuclear magnetic resonance (NMR) spectroscopy revealed that polyvinylpyrrolidone (PVP), polyvinylpyrrolidone/vinyl acetate (PVP-VA), and hypromellose (HPMC) distributed into the ibuprofen (IBP)-rich phase formed by LLPS when the amorphous solubility of IBP was exceeded. The amount of polymer in the drug-rich phase increased for higher-molecular-weight grades of PVP and HPMC. Moreover, PVP-VA showed a greater extent of distribution into the IBP-rich phase compared to PVP, and this is attributed to its reduced hydrophilicity resulting from the incorporation of vinyl acetate monomers. Direct quantification by NMR measurements indicated that the IBP concentration in the aqueous phase decreased as the amount of polymer in the IBP-rich phase increased. This can be attributed to a reduction of the chemical potential of IBP in the IBP-rich phase. The reduction in dissolved IBP concentration was greater for the IBP/PVP-VA system compared to the IBP/HPMC system, as a result of more extensive drug-polymer interactions in the former system. The present study highlights the impact of polymer selection on the attainable supersaturation of the drug and the factors that need to be considered in the formulation of ASDs to obtain optimized in vivo performance.
Collapse
Affiliation(s)
- Keisuke Ueda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan.,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
| | - Vaibhav Pathak
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
29
|
Wilson VR, Mugheirbi NA, Mosquera-Giraldo LI, Deac A, Moseson DE, Smith DT, Novo DC, Borca CH, Slipchenko LV, Edgar KJ, Taylor LS. Interaction of Polymers with Enzalutamide Nanodroplets-Impact on Droplet Properties and Induction Times. Mol Pharm 2021; 18:836-849. [PMID: 33539105 DOI: 10.1021/acs.molpharmaceut.0c00833] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Amorphous solid dispersions (ASDs), which consist of a drug dispersed in a polymeric matrix, are increasingly being applied to improve the in vivo performance of poorly water-soluble drugs delivered orally. The polymer is a critical component, playing several roles including facilitating drug release from the ASD, as well as delaying crystallization from the supersaturated solution generated upon dissolution. Certain ASD formulations dissolve to produce amorphous drug-rich nanodroplets. The interaction of the polymer with these nanodroplets is poorly understood but is thought to be important for inhibiting crystallization in these systems. In this study, the impact of ionic polymers on the crystallization kinetics of enzalutamide from supersaturated solutions containing different amounts of amorphous nanodroplets was evaluated by determination of nucleation induction times. The amount of the polymer associated with the drug nanodroplets was also determined. When comparing two polymers, hydroxypropylmethyl cellulose acetate succinate (HPMCAS) and Eudragit E PO, it was found that the crystallization tendency and physical properties of the drug nanodroplets varied in the presence of these two polymers. Both polymers distributed between the aqueous phase and the drug-rich nanodroplets. A greater amount of Eudragit E PO was associated with the drug-rich nanodroplets. Despite this, Eudragit E PO was a less-effective crystallization inhibitor than HPMCAS in systems containing nanodroplets. In conclusion, in supersaturated solutions containing amorphous nanodroplets, the extent of association of a polymer with the drug nanodroplet does not solely predict crystallization inhibition.
Collapse
Affiliation(s)
- Venecia R Wilson
- Department of Industrial & Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Naila A Mugheirbi
- Department of Industrial & Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Laura I Mosquera-Giraldo
- Department of Industrial & Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Alexandru Deac
- Department of Industrial & Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Dana E Moseson
- Department of Industrial & Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Daniel T Smith
- Department of Industrial & Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Diana C Novo
- Department of Chemistry, College of Science, Virginia Tech, 1040 Drillfield Drive, Blacksburg, Virginia 24061, United States
| | - Carlos H Borca
- Department of Chemical and Biological Engineering, School of Engineering and Applied Science, Princeton University, 41 Olden Street, Princeton, New Jersey 08544, United States
| | - Lyudmila V Slipchenko
- Department of Chemistry, College of Science, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| | - Kevin J Edgar
- Department of Chemistry, College of Science, Virginia Tech, 1040 Drillfield Drive, Blacksburg, Virginia 24061, United States
| | - Lynne S Taylor
- Department of Industrial & Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|
30
|
Ueda K, Taylor LS. Partitioning of surfactant into drug-rich nanodroplets and its impact on drug thermodynamic activity and droplet size. J Control Release 2021; 330:229-243. [DOI: 10.1016/j.jconrel.2020.12.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/16/2020] [Accepted: 12/13/2020] [Indexed: 02/08/2023]
|
31
|
Zhang W, Noland R, Chin S, Petkovic M, Zuniga R, Santarra B, Conklin B, Hou HH, Nagapudi K, Gruenhagen JA, Yehl P, Chen T. Impact of polymer type, ASD loading and polymer-drug ratio on ASD tablet disintegration and drug release. Int J Pharm 2021; 592:120087. [PMID: 33189812 DOI: 10.1016/j.ijpharm.2020.120087] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 10/23/2022]
Abstract
Amorphous solid dispersion (ASD) has become an attractive strategy to enhance solubility and bioavailability of poorly water-soluble drugs. To facilitate oral administration, ASDs are commonly incorporated into tablets. Disintegration and drug release from ASD tablets are thus critical for achieving the inherent solubility advantage of amorphous drugs. In this work, the impact of polymer type, ASD loading in tablet and polymer-drug ratio in ASD on disintegration and drug release of ASD tablets was systematically studied. Two hydrophilic polymers PVPVA and HPMC and one relatively hydrophobic polymer HPMCAS were evaluated. Dissolution testing was performed, and disintegration time was recorded during dissolution testing. As ASD loading increased, tablet disintegration time increased for all three polymer-based ASD tablets, and this effect was more pronounced for hydrophilic polymer-based ASD tablets. As polymer-drug ratio increased, tablet disintegration time increased for hydrophilic polymer-based ASD tablets, however, it remained short and largely unchanged for HPMCAS-based ASD tablets. Consequently, at high ASD loadings or high polymer-drug ratios, HPMCAS-based ASD tablets showed faster drug release than PVPVA- or HPMC-based ASD tablets. These results were attributed to the differences between polymer hydrophilicities and viscosities of polymer aqueous solutions. This work is valuable for understanding the disintegration and drug release of ASD tablets and provides insight to ASD composition selection from downstream tablet formulation perspective.
Collapse
Affiliation(s)
- Wei Zhang
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Ryan Noland
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Steven Chin
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Milan Petkovic
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ruth Zuniga
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Bethany Santarra
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Breanna Conklin
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hao Helen Hou
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Karthik Nagapudi
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jason A Gruenhagen
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Peter Yehl
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Tao Chen
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
32
|
Nichols BLB, Chen J, Mischnick P, Edgar KJ. Selective Oxidation of 2-Hydroxypropyl Ethers of Cellulose and Dextran: Simple and Efficient Introduction of Versatile Ketone Groups to Polysaccharides. Biomacromolecules 2020; 21:4835-4849. [PMID: 33236636 DOI: 10.1021/acs.biomac.0c01045] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Oxidation of polysaccharides has been a useful approach to new materials. However, selectivity in oxidation of polysaccharide macromolecular polyols remains a significant challenge with few methods for the synthesis of ketone-substituted polysaccharides. We report here a selective, practical, and efficient process, beginning with 2-hydroxypropyl ethers of polysaccharides that are simple and economical to prepare. We demonstrate this approach herein using commercial 2-hydroxypropyl cellulose (HPC) and 2-hydroxypropyl dextran (HPD) that we prepared. We oxidize the terminal, secondary alcohols of the oligo(2-hydroxypropyl) substituents with sodium hypochlorite so that the product has an oligo(2-hydroxypropyl) side chains terminated by a ketone. We demonstrate the high chemo- and regioselectivity of this oxidation by analytical methods including hydrolysis to monosaccharides and mass spectrometry of the resulting mixture. We provide an initial demonstration of the potential utility of these keto-polysaccharides by reacting Ox-HPC with primary amines to form Schiff base imines, providing proactive polymers.
Collapse
Affiliation(s)
- Brittany L B Nichols
- Department of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Junyi Chen
- Department of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Petra Mischnick
- Institute of Food Chemistry, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Kevin J Edgar
- Department of Sustainable Biomaterials, Virginia Tech, Blacksburg, Virginia 24061, United States.,Macromolecules Innovation Institute, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
33
|
Wilson VR, Lou X, Osterling DJ, Stolarik DF, Jenkins GJ, Nichols BLB, Dong Y, Edgar KJ, Zhang GGZ, Taylor LS. Amorphous solid dispersions of enzalutamide and novel polysaccharide derivatives: investigation of relationships between polymer structure and performance. Sci Rep 2020; 10:18535. [PMID: 33116200 PMCID: PMC7595150 DOI: 10.1038/s41598-020-75077-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 09/04/2020] [Indexed: 11/09/2022] Open
Abstract
Amorphous solid dispersion (ASD) is a widely employed formulation technique for drugs with poor aqueous solubility. Polymers are integral components of ASDs, but mechanisms by which polymers lead to the generation and maintenance of supersaturated solutions, which enhance oral absorption in vivo, are poorly understood. Herein, a diverse group of newly synthesized cellulose derivatives was evaluated for their ability to inhibit crystallization of enzalutamide, a poorly soluble compound used to treat prostate cancer. ASDs were prepared from selected polymers, specifically a somewhat hydrophobic polymer that was extremely effective at inhibiting drug crystallization, and a less effective, but more hydrophilic, crystallization inhibitor, that might afford better release. Drug membrane transport rate was evaluated in vitro and compared to in vivo performance, following oral dosing in rats. Good correlation was noted between the in vitro diffusion cell studies and the in vivo data. The ASD formulated with the less effective crystallization inhibitor outperformed the ASD prepared with the highly effective crystallization inhibitor in terms of the amount and rate of drug absorbed in vivo. This study provides valuable insight into key factors impacting oral absorption from enabling ASD formulations, and how best to evaluate such formulations using in vitro approaches.
Collapse
Affiliation(s)
- Venecia R Wilson
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, Lafayette, IN, 47907, USA
| | - Xiaochun Lou
- Drug Product Development, Research & Development, AbbVie, 1 N. Waukegan Road, North Chicago, IL, 60064, USA
| | - Donald J Osterling
- Drug Metabolism and Pharmacokinetics, Research & Development, AbbVie, 1 N. Waukegan Road, North Chicago, IL, 60064, USA
| | - DeAnne F Stolarik
- Drug Metabolism and Pharmacokinetics, Research & Development, AbbVie, 1 N. Waukegan Road, North Chicago, IL, 60064, USA
| | - Gary J Jenkins
- Drug Metabolism and Pharmacokinetics, Research & Development, AbbVie, 1 N. Waukegan Road, North Chicago, IL, 60064, USA
| | - Brittany L B Nichols
- Department of Chemistry, College of Science, Virginia Tech, 240 Kelly Hall, Blacksburg, VA, 24061, USA
| | - Yifan Dong
- Department of Chemistry, College of Science, Virginia Tech, 240 Kelly Hall, Blacksburg, VA, 24061, USA
| | - Kevin J Edgar
- Department of Sustainable Biomaterials, Virginia Tech, 230A Cheatham Hall, Blacksburg, VA, 24061, USA
| | - Geoff G Z Zhang
- Drug Product Development, Research & Development, AbbVie, 1 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, Lafayette, IN, 47907, USA.
| |
Collapse
|
34
|
Sahoo A, Suryanarayanan R, Siegel RA. Stabilization of Amorphous Drugs by Polymers: The Role of Overlap Concentration (C*). Mol Pharm 2020; 17:4401-4406. [DOI: 10.1021/acs.molpharmaceut.0c00576] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
35
|
Li N, Cape JL, Mankani BR, Zemlyanov DY, Shepard KB, Morgen MM, Taylor LS. Water-Induced Phase Separation of Spray-Dried Amorphous Solid Dispersions. Mol Pharm 2020; 17:4004-4017. [PMID: 32931293 PMCID: PMC7539301 DOI: 10.1021/acs.molpharmaceut.0c00798] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
![]()
Spray
drying is widely used in the manufacturing of amorphous solid
dispersion (ASD) systems due to its fast drying rate, enabling kinetic
trapping of the drug in amorphous form. Spray-drying conditions, such
as solvent composition, can have a profound impact on the properties
of spray-dried dispersions. In this study, the phase behavior of spray-dried
dispersions from methanol and methanol–water mixtures was assessed
using ritonavir and copovidone [poly(vinylpyrrolidone-co-vinyl acetate)
(PVPVA)] as dispersion components. The resultant ASDs were characterized
using differential scanning calorimetry (DSC), fluorescence spectroscopy,
X-ray photoelectron spectroscopy (XPS), as well as surface-normalized
dissolution rate (SNDR) measurements. Quaternary phase diagrams were
calculated using a four-component Flory–Huggins model. It was
found that the addition of water to the solvent system can lead to
phase separation during the spray-drying process. A 10:90 H2O/MeOH solvent system caused a minor extent of phase separation.
Phase heterogeneity in the 50 and 75% drug loading ASDs prepared from
this spray solvent can be detected using DSC but not with other techniques
used. The 25% drug loading system did not show phase heterogeneity
in solid-state characterization but exhibited a compromised dissolution
rate compared to that of the miscible ASD prepared from H2O-free solvent. This is possibly due to the formation of slow-releasing
drug-rich phases upon phase separation. ASDs prepared with a 60:40
H2O/MeOH solvent mixture showed phase heterogeneity with
all analytical methods used. The surface composition of dispersion
particles as measured by fluorescence spectroscopy and XPS showed
good agreement, suggesting surface drug enrichment of the spray-dried
ASD particles prepared from this solvent system. Calculated phase
diagrams and drying trajectories were consistent with experimental
observations, suggesting that small variations in solvent composition
may cause significant changes in ASD phase behavior during drying.
These findings should aid in spray-drying process development for
ASD manufacturing and can be applied broadly to assess the risk of
phase separation for spray-drying systems using mixed organic solvents
or other solvent-based processes.
Collapse
Affiliation(s)
- Na Li
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States.,Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road Unit 3092, Storrs, Connecticut 06269, United States
| | - Jonathan L Cape
- Research & Development, Lonza Pharma and Biotech, 1201 NW Wall Street, Suite 200, Bend, Oregon 97703, United States
| | - Bharat R Mankani
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States.,MarqMetrix Inc., Emerald Landing, 2157 N Northlake Way #240, Seattle, Washington 98103, United States
| | - Dmitry Y Zemlyanov
- Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana 47907, United States
| | - Kimberly B Shepard
- Research & Development, Lonza Pharma and Biotech, 1201 NW Wall Street, Suite 200, Bend, Oregon 97703, United States
| | - Michael M Morgen
- Research & Development, Lonza Pharma and Biotech, 1201 NW Wall Street, Suite 200, Bend, Oregon 97703, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|
36
|
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]
|
37
|
Trasi NS, Bhujbal SV, Zemlyanov DY, Zhou QT, Taylor LS. Physical stability and release properties of lumefantrine amorphous solid dispersion granules prepared by a simple solvent evaporation approach. INTERNATIONAL JOURNAL OF PHARMACEUTICS-X 2020; 2:100052. [PMID: 32760909 PMCID: PMC7390794 DOI: 10.1016/j.ijpx.2020.100052] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 07/07/2020] [Accepted: 07/12/2020] [Indexed: 11/26/2022]
Abstract
Amorphous solid dispersions (ASDs) of lumefantrine, which has low aqueous solubility, have been shown to improve bioavailability relative to crystalline formulations. Herein, the crystallization tendency and release properties of a variety of lumefantrine ASD granules, formed on a blend of microcrystalline cellulose and anhydrous lactose, prepared using a simple solvent evaporation method, were evaluated. Several polymers, a majority of which contained acidic moieties, and different drug loadings were assessed. Crystallinity as a function of time following exposure to stress storage conditions of 40 °C and 75% relative humidity was monitored for the various dispersions. Release testing was performed and ASD characteristics were further evaluated using infrared and X-ray photoelectron spectroscopy (XPS). A large difference in stability to crystallization was observed between the various ASDs, most notably depending on polymer chemistry. This could be largely rationalized based on the extent of drug-polymer interactions, specifically the degree of lumefantrine-polymer salt formation, which could be readily assessed with XPS spectroscopy. Lumefantrine release from the ASDs also varied considerably, whereby the best polymer for promoting physical stability did not lead to the highest extent of drug release. Several formulations led to concentrations above the amorphous solubility of lumefantrine, with the formation of nano-sized drug-rich aggregates. A balance between the ability of a given polymer to promote physical stability and drug release may need to be sought.
Collapse
Affiliation(s)
- Niraj S Trasi
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, USA
| | - Sonal V Bhujbal
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, USA
| | - Dmitry Y Zemlyanov
- Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana 47907, USA
| | - Qi Tony Zhou
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, USA
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, USA
| |
Collapse
|
38
|
Schittny A, Huwyler J, Puchkov M. Mechanisms of increased bioavailability through amorphous solid dispersions: a review. Drug Deliv 2020; 27:110-127. [PMID: 31885288 PMCID: PMC6968646 DOI: 10.1080/10717544.2019.1704940] [Citation(s) in RCA: 192] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Amorphous solid dispersions (ASDs) can increase the oral bioavailability of poorly soluble drugs. However, their use in drug development is comparably rare due to a lack of basic understanding of mechanisms governing drug liberation and absorption in vivo. Furthermore, the lack of a unified nomenclature hampers the interpretation and classification of research data. In this review, we therefore summarize and conceptualize mechanisms covering the dissolution of ASDs, formation of supersaturated ASD solutions, factors responsible for solution stabilization, drug uptake from ASD solutions, and drug distribution within these complex systems as well as effects of excipients. Furthermore, we discuss the importance of these findings on the development of ASDs. This improved overall understanding of these mechanisms will facilitate a rational ASD formulation development and will serve as a basis for further mechanistic research on drug delivery by ASDs.
Collapse
Affiliation(s)
- Andreas Schittny
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, Basel, Switzerland.,Department of Biomedicine, Division of Clinical Pharmacology and Toxicology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Jörg Huwyler
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, Basel, Switzerland
| | - Maxim Puchkov
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, Basel, Switzerland
| |
Collapse
|
39
|
Zhang Q, Ren W, Dushkin AV, Su W. Preparation, characterization, in vitro and in vivo studies of olmesartan medoxomil in a ternary solid dispersion with N-methyl-D-glucamine and hydroxypropyl-β-cyclodextrin. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101546] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
40
|
Saboo S, Kestur US, Flaherty DP, Taylor LS. Congruent Release of Drug and Polymer from Amorphous Solid Dispersions: Insights into the Role of Drug-Polymer Hydrogen Bonding, Surface Crystallization, and Glass Transition. Mol Pharm 2020; 17:1261-1275. [DOI: 10.1021/acs.molpharmaceut.9b01272] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Sugandha Saboo
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Umesh S. Kestur
- Drug Product Science and Technology, Bristol-Myers Squibb Company, One Squib Drive, New Brunswick, New Jersey 08903, United States
| | - Daniel P. Flaherty
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Lynne S. Taylor
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|
41
|
Ueda K, Taylor LS. Polymer Type Impacts Amorphous Solubility and Drug-Rich Phase Colloidal Stability: A Mechanistic Study Using Nuclear Magnetic Resonance Spectroscopy. Mol Pharm 2020; 17:1352-1362. [PMID: 32097023 DOI: 10.1021/acs.molpharmaceut.0c00061] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The polymer used in an amorphous solid dispersion (ASD) formulation plays a critical role in dosage form performance. Herein, drug-polymer interactions in aqueous solution were evaluated in order to better understand the dispersion stability of the colloidal drug-rich phase generated when the amorphous solubility is exceeded. The amorphous solubility (Sa,IBP) of ibuprofen (IBP) decreased when hypromellose (HPMC) or polyvinylpyrrolidone/vinyl acetate (PVP-VA) were present in solution. Solution nuclear magnetic resonance (NMR) spectroscopy revealed that a large amount of HPMC and PVP-VA distributed into the IBP-rich phase. The mixing of HPMC and PVP-VA with the IBP-rich phase led to the decreased Sa,IBP. In contrast, the charged amino methacrylate copolymer (Eudragit E PO, EUD-E) showed minimal mixing with the IBP-rich phase; however, this polymer did lead to solubilization of IBP in the bulk aqueous phase. Although the IBP-rich phase generated by dissolving IBP at concentrations above Sa,IBP rapidly coarsened followed by creaming in the absence of polymer, all of the polymers stabilized the IBP dispersion to some extent. The droplet size of the IBP-rich phase immediately after formation was around 300 nm in HPMC and PVP-VA solutions, and around 800 nm in the EUD-E solution. The mixing of the former two polymers with the drug-rich phase is thought to account for the smaller droplet size. Despite a smaller initial size, the dispersion stability of the IBP-rich droplets was relatively poor in the presence of PVP-VA. In contrast, the coalescence of the IBP-rich droplets was effectively suppressed by the steric repulsion and electrostatic repulsion derived from adsorbed HPMC and EUD-E, respectively. The present study highlights the complex effects of a polymer on the drug amorphous solubility and colloidal stability, which should be considered when optimizing ASD formulations with the goal of maximizing drug absorption.
Collapse
Affiliation(s)
- Keisuke Ueda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan.,Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
42
|
Schittny A, Philipp-Bauer S, Detampel P, Huwyler J, Puchkov M. Mechanistic insights into effect of surfactants on oral bioavailability of amorphous solid dispersions. J Control Release 2020; 320:214-225. [PMID: 31978445 DOI: 10.1016/j.jconrel.2020.01.031] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/05/2019] [Accepted: 01/17/2020] [Indexed: 02/04/2023]
Abstract
Drug delivery of poorly soluble drugs in form amorphous solid dispersions (ASDs) is an appealing method to increase in vivo bioavailability. For rational formulation design, a mechanistic understanding of the impact of surfactants on the performance of ASD-based formulations is therefore of importance. In this study, we used hot-melt extrusion to prepare ASDs composed of the model drug substance efavirenz with hydroxypropyl methylcellulose phthalate (HPMCP) as the base polymer, and surfactants. Molecular dynamics simulations and in vitro dissolution studies were used to investigate formation and drug release from polymer vesicles, and their ability to maintain a supersaturation state as a function of surfactant composition. It was possible to identify main factors regulating particle formation and to modify dissolution profiles with different excipient compositions. Animal studies in the rat, in combination with physiologically based pharmacokinetic modeling, demonstrated enhanced drug absorption from formed vesicles. The surfactant composition in the ASD had a direct influence on the morphology of these vesicles, as well as kinetics of drug release, and, therefore, the oral bioavailability. ASDs, prepared by hot-melt extrusion method, were optimized for dissolution and adsorption rates increase. Our findings contribute to a better understanding of dissolution behavior of ASDs with respect to the function of surfactants, aiming to facilitate a rational formulation development and an accelerated transition from in vitro systems to in vivo applications.
Collapse
Affiliation(s)
- A Schittny
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Switzerland; Division of Clinical Pharmacology and Toxicology, Department of Biomedicine, University Hospital Basel and University of Basel, Switzerland
| | - S Philipp-Bauer
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Switzerland
| | - P Detampel
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Switzerland
| | - J Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Switzerland
| | - M Puchkov
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Switzerland.
| |
Collapse
|
43
|
Trasi NS, Bhujbal S, Zhou QT, Taylor LS. Amorphous solid dispersion formation via solvent granulation - A case study with ritonavir and lopinavir. INTERNATIONAL JOURNAL OF PHARMACEUTICS-X 2019; 1:100035. [PMID: 31788669 PMCID: PMC6880113 DOI: 10.1016/j.ijpx.2019.100035] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/28/2019] [Accepted: 09/21/2019] [Indexed: 12/14/2022]
Abstract
Herein, we evaluate the potential of using a simple solvent granulation process to prepare a binary drug amorphous solid dispersion (ASD) containing two anti-HIV drugs, ritonavir and lopinavir. The drugs were granulated onto a mixture of lactose and microcrystalline cellulose, followed by drying to remove the solvent. The resultant granules were characterized and each drug was found to be X-ray amorphous. No crystallization was observed following storage for 1 month under accelerated stability conditions (40 °C and 75% relative humidity). The dissolution behavior of the compacted granules was compared with the marketed formulation. The dissolution rate of ritonavir was found to be significantly retarded relative to the commercial product when the two drugs were co-granulated. However, comparable release could be achieved when each drug was individually granulated, followed by combination and compaction. The solvent granulation approach may be a viable method to make ASDs of low dose drugs with low crystallization tendencies.
Collapse
Affiliation(s)
| | | | | | - Lynne S. Taylor
- Corresponding author at: Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, USA.
| |
Collapse
|
44
|
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
|
45
|
Han YR, Ma Y, Lee PI. Impact of phase separation morphology on release mechanism of amorphous solid dispersions. Eur J Pharm Sci 2019; 136:104955. [PMID: 31199980 DOI: 10.1016/j.ejps.2019.104955] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/09/2019] [Accepted: 06/10/2019] [Indexed: 11/26/2022]
Abstract
Amorphous solid dispersions (ASDs) can phase separate in the gel phase during dissolution, lowering the chemical potential and thus the driving force for drug release. The purpose of this study is to explore the connection between amorphous phase separation in the hydrated ASD and its resulting release rate. Poorly soluble model compounds - indomethacin (IND) and ritonavir (RTV) - were formulated as ASDs using PVP as carrier. Rotating disk dissolution studies with varying drug loading levels of IND-PVP and RTV-PVP showed that the drug release was fastest at an intermediate drug loading level. This was in part due to faster erosion of the ASD at lower drug loading levels. More interestingly, at low drug loading levels, PVP and the drug co-eroded, while at high drug loading levels, PVP was released preferentially. In the case of RTV-PVP, the loading level corresponding to this transition was correlated with the change in phase separation morphology as probed by confocal fluorescence imaging studies. At low drug loading levels, the hydrophobic domains were discrete domains while at high drug loading levels, hydrophobic domains were continuous. Our results suggest that at low drug loadings, release is mediated by erosion of the polymer along with embedded drug rich droplets, whereas at high drug loadings, formation of a drug-rich domain continuous morphology leads to preferential release of the polymer-rich domains. The transition from hydrophobic discrete to hydrophobic continuous morphology occurs at the percolation threshold. We discuss the two mechanisms of phase separation and its impact on the drug release from ASDs in the context of the ternary phase diagram.
Collapse
Affiliation(s)
- Yi Rang Han
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Yingshan Ma
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Ping I Lee
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada.
| |
Collapse
|
46
|
Guan J, Huan X, Liu Q, Jin L, Wu H, Zhang X, Mao S. Synergetic effect of nucleation and crystal growth inhibitor on in vitro-in vivo performance of supersaturable lacidipine solid dispersion. Int J Pharm 2019; 566:594-603. [PMID: 31175988 DOI: 10.1016/j.ijpharm.2019.06.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/03/2019] [Accepted: 06/05/2019] [Indexed: 10/26/2022]
Abstract
Limited supersaturation maintaining duration is the main challenge for amorphous solid dispersion design. Nucleation or crystal growth inhibitors may function in different ways but the combination use of nucleation and crystal growth inhibitors in supersaturated system is rarely explored. Thus, using Lacidipine (LCDP) as a Biopharmaceutical Classification System (BCS) II model drug, the aim of this study was to explore whether the combination use of nucleation and crystal growth inhibitors could provide a synergistic effect on the in vitro-in vivo performance of poorly water-soluble drugs. First of all, based on compatibility screening using solubility parameter (Δδ) and crystallization inhibition efficiency as criteria, soluplus (SOL) and gum arabic (GA) were selected as the most effective nucleation and crystal growth inhibitor respectively. Thereafter, the supersaturated drug solutions were spray dried and characterized. The in vitro release, physical stability as well as pharmacokinetic behavior were investigated. It was found that the combination use of SOL and GA did not present remarkable advantage in prolonging the supersaturation time in solution state. However, their synergistic effect in equilibrium solubility and dissolution enhancement was noticed at SOL/GA ratio 3:1, with 5-7 times higher dissolution rate observed for LCDP/SOL/GA based formulation compared with that of LCDP/SOL, which was maintained even after three months accelerated stability test under non-sink condition. Moreover, compared to the LCDP/SOL formulation, approximately 2.8 and 2.5-fold increase in the maximum plasma concentration (Cmax) and the area under the plasma-time curve (AUC0-∞) was achieved with LCDP/SOL/GA based formulation. Possible mechanism of the synergistic effect was elucidated, indicating GA may penetrate into SOL particles providing both electrostatic and steric stabilization. In conclusion, the combination use of screened nucleation and crystal growth inhibitors might be an efficient approach to design supersaturated drug delivery system.
Collapse
Affiliation(s)
- Jian Guan
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xu Huan
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qiaoyu Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Liwei Jin
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Haiyang Wu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xin Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shirui Mao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
47
|
Ishizuka Y, Ueda K, Okada H, Takeda J, Karashima M, Yazawa K, Higashi K, Kawakami K, Ikeda Y, Moribe K. Effect of Drug–Polymer Interactions through Hypromellose Acetate Succinate Substituents on the Physical Stability on Solid Dispersions Studied by Fourier-Transform Infrared and Solid-State Nuclear Magnetic Resonance. Mol Pharm 2019; 16:2785-2794. [DOI: 10.1021/acs.molpharmaceut.9b00301] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Yuya Ishizuka
- 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
| | - Hitomi Okada
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Junpei Takeda
- Analytical Development, Pharmaceutical Sciences, Takeda Pharmaceutical Company Limited, 2-26-1, Muraoka-Higashi, Fujisawa 251-8555, Kanagawa, Japan
| | - Masatoshi Karashima
- Analytical Development, Pharmaceutical Sciences, Takeda Pharmaceutical Company Limited, 2-26-1, Muraoka-Higashi, Fujisawa 251-8555, Kanagawa, Japan
| | - Koji Yazawa
- JEOL Resonance Incorpation, 3-1-2 Musashino, Akishima 196-8558, Tokyo, Japan
| | - Kenjirou Higashi
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Kohsaku Kawakami
- International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba 305-0044, Ibaraki, Japan
| | - Yukihiro Ikeda
- Analytical Development, Pharmaceutical Sciences, Takeda Pharmaceutical Company Limited, 2-26-1, Muraoka-Higashi, Fujisawa 251-8555, Kanagawa, Japan
| | - Kunikazu Moribe
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| |
Collapse
|
48
|
Saboo S, Mugheirbi NA, Zemlyanov DY, Kestur US, Taylor LS. Congruent release of drug and polymer: A “sweet spot” in the dissolution of amorphous solid dispersions. J Control Release 2019; 298:68-82. [DOI: 10.1016/j.jconrel.2019.01.039] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 11/29/2022]
|
49
|
Microstructure Formation for Improved Dissolution Performance of Lopinavir Amorphous Solid Dispersions. Mol Pharm 2019; 16:1751-1765. [PMID: 30811205 DOI: 10.1021/acs.molpharmaceut.9b00117] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
50
|
Hate SS, Reutzel-Edens SM, Taylor LS. Insight into Amorphous Solid Dispersion Performance by Coupled Dissolution and Membrane Mass Transfer Measurements. Mol Pharm 2018; 16:448-461. [PMID: 30521350 DOI: 10.1021/acs.molpharmaceut.8b01117] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The tendency of highly supersaturated solutions of poorly water-soluble drugs to undergo liquid-liquid phase separation (LLPS) into drug-rich and water-rich phases when the concentration exceeds the amorphous solubility, for example, during dissolution of some amorphous solid dispersions, is thought to be advantageous from a bioavailability enhancement perspective. Recently, we have developed a high surface area, flow-through absorptive dissolution testing apparatus that enables fast mass transfer providing more in vivo relevant conditions and time frames for formulation testing. Using this apparatus, the absorption behaviors of solutions with different extents of supersaturation below and above the amorphous solubility were evaluated. In addition, simultaneous dissolution-absorption testing of amorphous solid dispersions (ASDs) with varying drug loadings and polymer types was carried out to study and distinguish the absorption behavior of ASDs that do or do not undergo LLPS. When compared with closed-compartment dissolution testing, a significant influence of the absorptive compartment on the dissolution rate of ASDs, particularly at high drug loadings, was observed. The formation of drug-rich nanodroplets, generated by both solvent-addition and ASD dissolution, resulted in a higher amount of drug transferred across the membrane. Moreover, the mass transfer was further enhanced with increasing concentration above the amorphous solubility, thereby showing correlation with an increase in the number of drug-rich particles. The importance of including an absorptive compartment in dissolution testing is highlighted in this study, enabling coupling of dissolution to membrane transport, and providing a more meaningful comparison between different formulations.
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
|