1
|
Han X, Kawakami K. Influence of Pore Size of Mesoporous Silica on Physical Stability of Overloaded Celecoxib Glass. Mol Pharm 2025; 22:2556-2567. [PMID: 40183771 DOI: 10.1021/acs.molpharmaceut.4c01482] [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: 04/05/2025]
Abstract
The stabilization mechanism of mesoporous silica (MS) of two different pore sizes (21 and 2.5 nm) on overloaded celecoxib (CEL) glass was investigated. Differential scanning calorimetry (DSC) measurements revealed the presence of three fractions with different molecular mobilities: free, intermediate, and rigid ones. The free fraction exhibited cold crystallization during DSC heating and was assumed to have almost the same properties as those of the bulk molecules. The rigid fraction did not exhibit either glass transition or cold crystallization behavior, which should be stabilized by interactions with the MS surface. The remaining molecules exhibited glass transition behavior without any tendency toward cold crystallization during heating, which is called the intermediate fraction. The molecular dynamics of each fraction was investigated by using broadband dielectric spectroscopy (BDS). While the intermediate and free fractions exhibited comparable mobility, the rigid fraction demonstrated pore-size-dependent behavior: enhanced and suppressed molecular mobility was observed for the rigid fraction confined in 21 and 2.5 nm-pores, respectively. Isothermal crystallization of CEL glass was investigated using DSC and BDS at 95 °C. The results revealed that the CEL glass mixed with MS with large pores exhibited slower crystallization compared to the CEL glass without MS, whereas accelerated crystallization was observed for the CEL mixed with a small amount of MS of small pores. The pore size of 21 nm was much larger than the cooperatively rearranging region (CRR) of the CEL glass, whereas the pore size of 2.5 nm was comparable to that. When the pore size was larger than that of the CRR, most of the loaded CEL molecules behaved as an intermediate fraction, presumably because the molecules could exchange inside and outside the pore. In contrast, the exchange was not likely to proceed when the pore size was comparable to or smaller than that of the CRR, leaving a large free fraction. This finding provides a deep understanding of the stabilization mechanism of overloaded pharmaceutical glass by using mesoporous materials.
Collapse
Affiliation(s)
- Xue Han
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
- Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Kohsaku Kawakami
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
- Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| |
Collapse
|
2
|
Kawakami K, Ohyama K. Crystal Nucleation in Ibuprofen Glass: Possible Relevance between the Characteristic Length of the Cooperatively Rearranging Region and the Size of Crystal Nuclei. J Phys Chem B 2025; 129:2096-2104. [PMID: 39915259 PMCID: PMC11848918 DOI: 10.1021/acs.jpcb.4c07005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/21/2025]
Abstract
Crystallization behavior of ibuprofen glass was investigated with focus on the nucleation process and its possible relevance to the cooperatively rearranging region (CRR). The nucleation temperature range of ibuprofen glass was determined by annealing it at various temperatures, followed by observation of the probability of cold crystallization. The temperature to provide the highest probability of nucleation was -15 °C. The effect of the addition of a polymer was also investigated to find that it enhanced and suppressed the crystallization depending on the polymer species and its amount added. The added polymer seemed to influence both nucleation and crystal growth processes by decreasing the glass/nuclei interfacial tension and increasing viscosity, respectively. In addition, the coincidence of the size of CRR in the presence of the polymer with the critical size of nuclei was assumed to enhance nucleation. This finding provides a novel viewpoint for clarifying the nucleation mechanism from supercooled liquids and glasses.
Collapse
Affiliation(s)
- Kohsaku Kawakami
- Research
Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki Tsukuba, Ibaraki 305-0044, Japan
- Graduate
School of Pure and Applied Sciences, University
of Tsukuba, 1-1-1 Tennodai
Tsukuba, Ibaraki 305-8577, Japan
| | - Kaoru Ohyama
- Research
Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki Tsukuba, Ibaraki 305-0044, Japan
| |
Collapse
|
3
|
Kawakami K, Ishitsuka T, Fukiage M, Nishida Y, Shirai T, Hirai Y, Hideshima T, Tanabe F, Shinoda K, Tamate R, Fujita T. Long-term physical stability of amorphous solid dispersions: Comparison of detection powers of common evaluation methods for spray-dried and hot-melt extruded formulations. J Pharm Sci 2025; 114:145-156. [PMID: 38950881 DOI: 10.1016/j.xphs.2024.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024]
Abstract
Although physical stability can be a critical issue during the development of amorphous solid dispersions (ASDs), there are no established protocols to predict/detect their physical stability. In this study, we have prepared fenofibrate ASDs using two representative manufacturing methods, hot-melt extrusion and spray-drying, to investigate their physical stability for one year. Intentionally unstable ASDs were designed to compare the detection power of each evaluation method, including X-ray powder diffraction (XRPD), differential scanning calorimetry (DSC), scanning electron microscopy (SEM), and dissolution study. Each method did not provide the same judgment results on physical stability in some cases because of their different evaluation principles and sensitivity, which has been well-comprehended only for one-component glass. This study revealed that the detection powers of each evaluation method significantly depended on the manufacturing methods. DSC was an effective method to detect a small amount of crystals for both types of ASDs in a quantitative manner. Although the sensitivity of XRPD was always lower compared to that of DSC, interpretation of the data was the easiest. SEM was very effective for observing the crystallization of the small amount of drug for hot-melt extruded products, as the drug crystal vividly appeared on the large grains. The dissolution performance of spray-dried products could change even without any indication of physical change including crystallization. The advantage/disadvantage and complemental roles of each evaluation method are discussed for deeper understanding on the physical stability data of ASDs.
Collapse
Affiliation(s)
- Kohsaku Kawakami
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan.
| | - Taichi Ishitsuka
- Pharmaceutical R&D, Ono Pharmaceutical Co., Ltd., 1-15-26, Kamiji, higashinari-ku, Osaka 537-0003, Japan
| | - Masafumi Fukiage
- Pharmaceutical R&D, Ono Pharmaceutical Co., Ltd., 1-15-26, Kamiji, higashinari-ku, Osaka 537-0003, Japan
| | - Yohei Nishida
- Sumitomo Pharma America, Inc., 84 Waterford Drive, Marlborough, MA 01752, USA
| | - Tetsuo Shirai
- API and Pharmaceutical Development Department, Fuji Chemical Industries Co., Ltd., 1, Gohkakizawa, Kamiichi, Nakaniikawa, Toyama 930-0397, Japan
| | - Yosuke Hirai
- API and Pharmaceutical Development Department, Fuji Chemical Industries Co., Ltd., 1, Gohkakizawa, Kamiichi, Nakaniikawa, Toyama 930-0397, Japan
| | - Tetsu Hideshima
- API and Pharmaceutical Development Department, Fuji Chemical Industries Co., Ltd., 1, Gohkakizawa, Kamiichi, Nakaniikawa, Toyama 930-0397, Japan
| | - Fumiaki Tanabe
- Nara Machinery Co., Ltd., 2-5-7 Jonan-Jima, Ohta-ku, Tokyo 143-0002, Japan
| | - Koji Shinoda
- Nara Machinery Co., Ltd., 2-5-7 Jonan-Jima, Ohta-ku, Tokyo 143-0002, Japan
| | - Ryota Tamate
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Takuya Fujita
- College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga 525-8577, Japan
| |
Collapse
|
4
|
Moreira GG, Taveira SF, Martins FT, Wagner KG, Marreto RN. Multivariate Analysis of Solubility Parameters for Drug-Polymer Miscibility Assessment in Preparing Raloxifene Hydrochloride Amorphous Solid Dispersions. AAPS PharmSciTech 2024; 25:127. [PMID: 38844724 DOI: 10.1208/s12249-024-02844-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/21/2024] [Indexed: 09/05/2024] Open
Abstract
The success of obtaining solid dispersions for solubility improvement invariably depends on the miscibility of the drug and polymeric carriers. This study aimed to categorize and select polymeric carriers via the classical group contribution method using the multivariate analysis of the calculated solubility parameter of RX-HCl. The total, partial, and derivate parameters for RX-HCl were calculated. The data were compared with the results of excipients (N = 36), and a hierarchical clustering analysis was further performed. Solid dispersions of selected polymers in different drug loads were produced using solvent casting and characterized via X-ray diffraction, infrared spectroscopy and scanning electron microscopy. RX-HCl presented a Hansen solubility parameter (HSP) of 23.52 MPa1/2. The exploratory analysis of HSP and relative energy difference (RED) elicited a classification for miscible (n = 11), partially miscible (n = 15), and immiscible (n = 10) combinations. The experimental validation followed by a principal component regression exhibited a significant correlation between the crystallinity reduction and calculated parameters, whereas the spectroscopic evaluation highlighted the hydrogen-bonding contribution towards amorphization. The systematic approach presented a high discrimination ability, contributing to optimal excipient selection for the obtention of solid solutions of RX-HCl.
Collapse
Affiliation(s)
- Guilherme G Moreira
- Laboratory of Nanosystems and Drug Delivery Devices (NanoSYS), School of Pharmacy, Universidade Federal de Goiás, Goiânia, Goiás, 74.605-170, Brazil
| | - Stephânia F Taveira
- Laboratory of Nanosystems and Drug Delivery Devices (NanoSYS), School of Pharmacy, Universidade Federal de Goiás, Goiânia, Goiás, 74.605-170, Brazil
| | - Felipe T Martins
- Institute of Chemistry, Universidade Federal de Goiás, Goiânia, 74.001-970, Brazil
| | - Karl G Wagner
- Department of Pharmaceutics, Pharmaceutical Institute, University of Bonn, 53121, Bonn, Germany
| | - Ricardo N Marreto
- Laboratory of Nanosystems and Drug Delivery Devices (NanoSYS), School of Pharmacy, Universidade Federal de Goiás, Goiânia, Goiás, 74.605-170, Brazil.
| |
Collapse
|
5
|
Han X, Dai K, Kawakami K. Influence of Nucleation on Relaxation, Molecular Cooperativity, and Physical Stability of Celecoxib Glass. Mol Pharm 2024; 21:1794-1803. [PMID: 38401048 DOI: 10.1021/acs.molpharmaceut.3c01116] [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: 02/26/2024]
Abstract
Although nucleation is considered the first step in the crystallization of glass materials, the structure and properties of the nuclei are not understood well. Influence of nucleation on the structure and dynamics of celecoxib glass was evaluated in this study. The nuclei for Form III were induced by annealing the glass at freezing temperature, and their impact on the relaxation behavior was investigated using thermal analysis and broadband dielectric spectroscopy to find accelerated α relaxation and suppressed β relaxation. In addition, observed after nucleation was a decrease in cooperativity of the molecular motion, presumably because of the appearance of void spaces in the glass structure. During long-term isothermal crystallization studies, crystal growth to Form III was accelerated in the presence of the nuclei, whereas this effect was less remarkable when a different crystal form dominated the crystallization behavior. These observations should provide more detailed insights into the nucleation mechanism and impact of nucleation on molecular dynamics including physical stability of pharmaceutical glasses. In addition, discussed is the remarkable acceleration of the crystallization rate of the celecoxib glass just below its Tg, which could be understood by diffusionless crystal growth.
Collapse
Affiliation(s)
- Xue Han
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Kexin Dai
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
- Department of Chemical Engineering, University of California, Santa Barbara, California 93106, United States
| | - Kohsaku Kawakami
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| |
Collapse
|
6
|
Svoboda R, Kozlová K. Thermo-Structural Characterization of Phase Transitions in Amorphous Griseofulvin: From Sub-T g Relaxation and Crystal Growth to High-Temperature Decomposition. Molecules 2024; 29:1516. [PMID: 38611796 PMCID: PMC11013327 DOI: 10.3390/molecules29071516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
The processes of structural relaxation, crystal growth, and thermal decomposition were studied for amorphous griseofulvin (GSF) by means of thermo-analytical, microscopic, spectroscopic, and diffraction techniques. The activation energy of ~395 kJ·mol-1 can be attributed to the structural relaxation motions described in terms of the Tool-Narayanaswamy-Moynihan model. Whereas the bulk amorphous GSF is very stable, the presence of mechanical defects and micro-cracks results in partial crystallization initiated by the transition from the glassy to the under-cooled liquid state (at ~80 °C). A key aspect of this crystal growth mode is the presence of a sufficiently nucleated vicinity of the disrupted amorphous phase; the crystal growth itself is a rate-determining step. The main macroscopic (calorimetrically observed) crystallization process occurs in amorphous GSF at 115-135 °C. In both cases, the common polymorph I is dominantly formed. Whereas the macroscopic crystallization of coarse GSF powder exhibits similar activation energy (~235 kJ·mol-1) as that of microscopically observed growth in bulk material, the activation energy of the fine GSF powder macroscopic crystallization gradually changes (as temperature and/or heating rate increase) from the activation energy of microscopic surface growth (~105 kJ·mol-1) to that observed for the growth in bulk GSF. The macroscopic crystal growth kinetics can be accurately described in terms of the complex mechanism, utilizing two independent autocatalytic Šesták-Berggren processes. Thermal decomposition of GSF proceeds identically in N2 and in air atmospheres with the activation energy of ~105 kJ·mol-1. The coincidence of the GSF melting temperature and the onset of decomposition (both at 200 °C) indicates that evaporation may initiate or compete with the decomposition process.
Collapse
Affiliation(s)
- Roman Svoboda
- Department of Physical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 532 10 Pardubice, Czech Republic;
| | | |
Collapse
|
7
|
Budiman A, Anastasya G, Handini AL, Lestari IN, Subra L, Aulifa DL. Characterization of Drug with Good Glass-Forming Ability Loaded Mesoporous Silica Nanoparticles and Its Impact Toward in vitro and in vivo Studies. Int J Nanomedicine 2024; 19:2199-2225. [PMID: 38465205 PMCID: PMC10924831 DOI: 10.2147/ijn.s453873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/18/2024] [Indexed: 03/12/2024] Open
Abstract
Solid oral dosage forms are mostly preferred in pharmaceutical formulation development due to patient convenience, ease of product handling, high throughput, low manufacturing costs, with good physical and chemical stability. However, 70% of drug candidates have poor water solubility leading to compromised bioavailability. This phenomenon occurs because drug molecules are often absorbed after dissolving in gastrointestinal fluid. To address this limitation, delivery systems designed to improve the pharmacokinetics of drug molecules are needed to allow controlled release and target-specific delivery. Among various strategies, amorphous formulations show significantly high potential, particularly for molecules with solubility-limited dissolution rates. The ease of drug molecules to amorphized is known as their glass-forming ability (GFA). Specifically, drug molecules categorized into class III based on the Taylor classification have a low recrystallization tendency and high GFA after cooling, with substantial "glass stability" when heated. In the last decades, the application of mesoporous silica nanoparticles (MSNs) as drug delivery systems (DDS) has gained significant attention in various investigations and the pharmaceutical industry. This is attributed to the unique physicochemical properties of MSNs, including high loading capacity, recrystallization inhibition, excellent biocompatibility, and easy functionalization. Therefore, this study aimed to discuss the current state of good glass former drug loaded mesoporous silica and shows its impact on the pharmaceutical properties including dissolution and physical stability, along with in vivo study. The results show the importance of determining whether mesoporous structures are needed in amorphous formulations to improve the pharmaceutical properties of drug with a favorable GFA.
Collapse
Affiliation(s)
- Arif Budiman
- Department of Pharmaceutics and Pharmaceutical Technology, Universitas Padjadjaran, Bandung, Indonesia
| | - Gracia Anastasya
- Department of Pharmaceutics and Pharmaceutical Technology, Universitas Padjadjaran, Bandung, Indonesia
| | - Annisa Luthfiyah Handini
- Department of Pharmaceutics and Pharmaceutical Technology, Universitas Padjadjaran, Bandung, Indonesia
| | - Ira Novianty Lestari
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Universitas Padjadjaran, Bandung, Indonesia
| | - Laila Subra
- Department of Pharmacy, Universiti Geomatika Malaysia, Kuala Lumpur, Malaysia
| | - Diah Lia Aulifa
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
8
|
Okada K, Ono T, Hayashi Y, Kumada S, Onuki Y. Use of Time-Domain NMR for 1H T 1 Relaxation Measurement and Fitting Analysis in Homogeneity Evaluation of Amorphous Solid Dispersion. J Pharm Sci 2024; 113:680-687. [PMID: 37659719 DOI: 10.1016/j.xphs.2023.08.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/04/2023]
Abstract
This study examined the usefulness of 1H T1 relaxation measurements for evaluating the homogeneity of amorphous solid dispersion (ASD). Indomethacin and polyvinylpyrrolidone were used to prepare two kinds of ASDs. One was inhomogeneous ASD (ASDmelt) prepared by a melt-quenching method, and the other was homogeneous ASD (ASDsolvent) prepared by a solvent evaporation method. The T1 relaxation was measured by the time-domain NMR (TD-NMR) technique using a low-field NMR system. Curve-fitting analysis of T1 relaxation plots was conducted using the Akaike information criterion. This fitting analysis revealed that the T1 relaxation of ASDmelt and ASDsolvent was biphasic and monophasic, respectively. ASDmelt and ASDsolvent were inhomogeneous and homogeneous on a nanometer scale, respectively, considering the spin diffusion of 1H nuclei. These T1 results were consistent with the Raman mapping of ASDs. From the fitting analysis of 1H T1 relaxation, we conclude that TD-NMR is a promising technique for evaluating ASD homogeneity.
Collapse
Affiliation(s)
- Kotaro Okada
- Laboratory of Pharmaceutical Technology, School of Pharmacy and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama 930-0194, Japan.
| | - Takashi Ono
- Laboratory of Pharmaceutical Technology, School of Pharmacy and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama 930-0194, Japan; Formulation Development Department, Development & Planning Division, Nichi-Iko Pharmaceutical Co., Ltd., 205-1 Shimoumezawa, Namerikawa-shi, Toyama 936-0857, Japan
| | - Yoshihiro Hayashi
- Formulation Development Department, Development & Planning Division, Nichi-Iko Pharmaceutical Co., Ltd., 205-1 Shimoumezawa, Namerikawa-shi, Toyama 936-0857, Japan
| | - Shungo Kumada
- Formulation Development Department, Development & Planning Division, Nichi-Iko Pharmaceutical Co., Ltd., 205-1 Shimoumezawa, Namerikawa-shi, Toyama 936-0857, Japan
| | - Yoshinori Onuki
- Laboratory of Pharmaceutical Technology, School of Pharmacy and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama 930-0194, Japan
| |
Collapse
|
9
|
Svoboda R, Macháčková J, Nevyhoštěná M, Komersová A. Thermal stability of amorphous nimesulide: from glass formation to crystal growth and thermal degradation. Phys Chem Chem Phys 2024; 26:856-872. [PMID: 38087904 DOI: 10.1039/d3cp02260a] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
Thermally induced physico-chemical transformations in amorphous nimesulide were studied by means of differential scanning calorimetry (DSC), thermogravimetry, and Raman microscopy. The equilibrium glass transition temperature was found to be Tg0 = 10-15 °C, and the relaxation motions were found to be temperature-dependent. Crystal growth from the amorphous phase was found to be crucially dependent on the presence of mechanical defects that serve as centers for heterogeneous nucleation. The large amounts of mechanical defects significantly decrease the activation energy of the macroscopic crystallization; the positions of the crystallization peaks and their asymmetry/shape remain however almost unchanged. At laboratory temperature, powdered nimesulide fully crystallizes within several hours, with an absolute majority of the crystalline phase being formed as the thermodynamically stable form I polymorph. Amorphous nimesulide does not crystallize from the free smooth surface (no trace of formed crystallites was found by optical microscopy after 30 days at laboratory temperature). Nimesulide was found to be very stable at temperatures above its melting point of 147.5 °C; thermal degradation starts to proceed slowly at 200 °C. Mutual correlations between the macroscopic and microscopic crystal growth processes and between the viscous flow and structural relaxation motions were discussed based on the values of the corresponding activation energies. A link between the cooperativity of structural domains, parameters of the Tool-Narayanaswamy-Moynihan relaxation model, and microscopic crystal growth was proposed.
Collapse
Affiliation(s)
- Roman Svoboda
- Department of Physical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 532 10 Pardubice, Czech Republic.
| | - Jana Macháčková
- Department of Physical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 532 10 Pardubice, Czech Republic.
| | - Marie Nevyhoštěná
- Department of Physical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 532 10 Pardubice, Czech Republic.
| | - Alena Komersová
- Department of Physical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 532 10 Pardubice, Czech Republic.
| |
Collapse
|
10
|
Ueda H, Hirakawa Y, Miyano T, Nakayama Y, Hatanaka Y, Uchiyama H, Tozuka Y, Kadota K. Improvement in Inhalation Properties of Theophylline and Levofloxacin by Co-Amorphization and Enhancement in Its Stability by Addition of Amino Acid as a Third Component. Mol Pharm 2023; 20:6368-6379. [PMID: 37942959 DOI: 10.1021/acs.molpharmaceut.3c00756] [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: 11/10/2023]
Abstract
Co-amorphous systems are amorphous formulations stabilized by the miscible dispersion of small molecules. This study aimed to design a stable co-amorphous system for the co-delivery of two drugs to the lungs as an inhaled formulation. Theophylline (THE) and levofloxacin (LEV) were used as model drugs for treating lung infection with inflammation. Leucine (LEU) or tryptophan (TRP) was employed as the third component to improve the inhalation properties. The co-amorphous system containing THE and LEV in an equal molar ratio was successfully prepared via spray drying where reduction of the particle size and change to the spherical morphology were observed. The addition of LEU or TRP at a one-tenth molar ratio to THE-LEV did not affect the formation of the co-amorphous system, but only TRP acted as an antiplasticizer. The Fourier transform infrared spectroscopy spectra revealed intermolecular interactions between THE and LEV in the co-amorphous system that were retained after the addition of LEU or TRP. The co-amorphous THE-LEV system exhibited better in vitro aerodynamic performance than a physical mixture of these compounds and permitted the simultaneous delivery of both drugs in various stages. The co-amorphous THE-LEV system crystallized at 40 °C, and this crystallization was not prevented by LEU. However, THE-LEV-TRP maintained its amorphous state for 1 month. Thus, TRP can act as a third component to improve the physical stability of the co-amorphous THE-LEV system, while maintaining the enhanced aerodynamic properties.
Collapse
Affiliation(s)
- Hiroshi Ueda
- Analysis and Evaluation Laboratory, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Yuya Hirakawa
- Analysis and Evaluation Laboratory, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Tetsuya Miyano
- Laboratory for Medicinal Chemistry Research, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Yuzuki Nakayama
- Department of Formulation Design and Pharmaceutical Technology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka 569-1094, Japan
| | - Yuta Hatanaka
- Department of Formulation Design and Pharmaceutical Technology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka 569-1094, Japan
| | - Hiromasa Uchiyama
- Department of Formulation Design and Pharmaceutical Technology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka 569-1094, Japan
| | - Yuichi Tozuka
- Department of Formulation Design and Pharmaceutical Technology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka 569-1094, Japan
| | - Kazunori Kadota
- Department of Formulation Design and Pharmaceutical Technology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka 569-1094, Japan
| |
Collapse
|
11
|
Jiang J, Lu A, Ma X, Ouyang D, Williams RO. The applications of machine learning to predict the forming of chemically stable amorphous solid dispersions prepared by hot-melt extrusion. Int J Pharm X 2023; 5:100164. [PMID: 36798832 PMCID: PMC9925947 DOI: 10.1016/j.ijpx.2023.100164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
Amorphous solid dispersion (ASD) is one of the most important strategies to improve the solubility and dissolution rate of poorly water-soluble drugs. As a widely used technique to prepare ASDs, hot-melt extrusion (HME) provides various benefits, including a solvent-free process, continuous manufacturing, and efficient mixing compared to solvent-based methods, such as spray drying. Energy input, consisting of thermal and specific mechanical energy, should be carefully controlled during the HME process to prevent chemical degradation and residual crystallinity. However, a conventional ASD development process uses a trial-and-error approach, which is laborious and time-consuming. In this study, we have successfully built multiple machine learning (ML) models to predict the amorphization of crystalline drug formulations and the chemical stability of subsequent ASDs prepared by the HME process. We utilized 760 formulations containing 49 active pharmaceutical ingredients (APIs) and multiple types of excipients. By evaluating the built ML models, we found that ECFP-LightGBM was the best model to predict amorphization with an accuracy of 92.8%. Furthermore, ECFP-XGBoost was the best in estimating chemical stability with an accuracy of 96.0%. In addition, the feature importance analyses based on SHapley Additive exPlanations (SHAP) and information gain (IG) revealed that several processing parameters and material attributes (i.e., drug loading, polymer ratio, drug's Extended-connectivity fingerprints (ECFP) fingerprints, and polymer's properties) are critical for achieving accurate predictions for the selected models. Moreover, important API's substructures related to amorphization and chemical stability were determined, and the results are largely consistent with the literature. In conclusion, we established the ML models to predict formation of chemically stable ASDs and identify the critical attributes during HME processing. Importantly, the developed ML methodology has the potential to facilitate the product development of ASDs manufactured by HME with a much reduced human workload.
Collapse
Affiliation(s)
- Junhuang Jiang
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Anqi Lu
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Xiangyu Ma
- Global Investment Research, Goldman Sachs, NY 10282, USA
| | - Defang Ouyang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, 999078, Macau
| | - Robert O. Williams
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
12
|
Moseson DE, Taylor LS. Crystallinity: A Complex Critical Quality Attribute of Amorphous Solid Dispersions. Mol Pharm 2023; 20:4802-4825. [PMID: 37699354 DOI: 10.1021/acs.molpharmaceut.3c00526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
Does the performance of an amorphous solid dispersion rely on having 100% amorphous content? What specifications are appropriate for crystalline content within an amorphous solid dispersion (ASD) drug product? In this Perspective, the origin and significance of crystallinity within amorphous solid dispersions will be considered. Crystallinity can be found within an ASD from one of two pathways: (1) incomplete amorphization, or (2) crystal creation (nucleation and crystal growth). While nucleation and crystal growth is the more commonly considered pathway, where crystals originate as a physical stability failure upon accelerated or prolonged storage, manufacturing-based origins of crystallinity are possible as well. Detecting trace levels of crystallinity is a significant analytical challenge, and orthogonal methods should be employed to develop a holistic assessment of sample properties. Probing the impact of crystallinity on release performance which may translate to meaningful clinical significance is inherently challenging, requiring optimization of dissolution test variables to address the complexity of ASD formulations, in terms of drug physicochemical properties (e.g., crystallization tendency), level of crystallinity, crystal reference material selection, and formulation characteristics. The complexity of risk presented by crystallinity to product performance will be illuminated through several case studies, highlighting that a one-size-fits-all approach cannot be used to set specification limits, as the risk of crystallinity can vary widely based on a multitude of factors. Risk assessment considerations surrounding drug physicochemical properties, formulation fundamentals, physical stability, dissolution, and crystal micromeritic properties will be discussed.
Collapse
Affiliation(s)
- Dana E Moseson
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
- Worldwide Research and Development Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
13
|
Miyazaki T, Mizoguchi R, Ueda K, Shinozaki T, Kamoto M, Takeda Y, Sakuma S, Ito N, Momo M, Kawakami K. Crystallization of Amorphous Nifedipine Under Isothermal Conditions: Inter-laboratory Reproducibility and Investigation of the Factors Affecting Reproducibility. J Pharm Sci 2023; 112:2703-2716. [PMID: 37301322 DOI: 10.1016/j.xphs.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023]
Abstract
High inter-laboratory reproducibility is required for conducting collaborative experiments among several laboratories. The primary aim of our evaluation of the physical stability of amorphous drugs, conducted in co-operation with eight laboratories, was to establish a protocol for isothermal storage tests to obtain data of the same quality from all the participating laboratories. Sharing a protocol that contained the same level of detail as the experimental section of general papers was insufficient for high inter-laboratory reproducibility. We investigated the causes of variations in the data from the various laboratories and restricted the protocol step-by-step to achieve high inter-laboratory reproducibility. The various experimentalists had very different levels of awareness regarding how to control the temperature of a sample as the samples were transferred into and out of thermostatic chambers. Specific instructions on how to conduct this operation, such as regarding the time required for the transfer and thermal protection of the container during the transfer, helped to reduce variation. Improved inter-laboratory reproducibility revealed that the physical stabilities of amorphous drugs differed when samples were prepared in differently shaped aluminum pans designed for various differential scanning calorimeters.
Collapse
Affiliation(s)
- Tamaki Miyazaki
- Division of Drugs, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan.
| | - Ryo Mizoguchi
- CMC Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Keisuke Ueda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba 260-8675, Japan
| | - Taeko Shinozaki
- Research Group V, Analytical & Quality Evaluation Research Laboratories, Daiichi Sankyo Co., Ltd., 1-12-1 Shinomiya, Hiratsuka, Kanagawa 254-0014, Japan
| | - Mie Kamoto
- Analytical Research Laboratories, Pharmaceutical Science & Technology Function Unit, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki 300-2635, Japan
| | - Yoshihiro Takeda
- Core Technology Research Department, X-ray Research Laboratory, Rigaku Corp., 3-9-12 Matsubara-cho, Akishima, Tokyo 196-8666, Japan
| | - Satoshi Sakuma
- Shionogi & Co., Ltd., 2-1-3 Kuise Terajima, Amagasaki, Hyogo 660-0813, Japan
| | - Naoya Ito
- Analytical Research & Development Laboratories, Sumitomo Pharma Co., Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka, 554-0022, Japan
| | - Michiko Momo
- Takeda Pharmaceutical Co., Ltd., 2-26-1 Muraokahigashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Kohsaku Kawakami
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| |
Collapse
|
14
|
Song J, Kawakami K. Nucleation During Storage Impeded Supersaturation in the Dissolution Process of Amorphous Celecoxib. Mol Pharm 2023; 20:4050-4057. [PMID: 37413788 DOI: 10.1021/acs.molpharmaceut.3c00210] [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: 07/08/2023]
Abstract
The aqueous solubility of active pharmaceutical ingredients (APIs) is one of the most critical factors in determining the absorption of orally administered drugs. Amorphization of API may offer better drug absorption than the crystalline state owing to enhanced solubility. However, if crystal nuclei are formed during storage, they may develop into crystals upon contact with water, thus limiting the dissolution advantage. In an earlier study, we found that the nuclei of amorphous celecoxib (CEL) could be formed at freezing temperatures (FT) without further crystal growth. Following this finding, we compared the dissolution performances of amorphous CEL annealed at room temperature (RT, 25 °C) or FT (-20 °C). We found that only the RT-annealed CEL could achieve a supersaturated state effectively during the dissolution process, which could be explained by the fast conversion of the FT-annealed amorphous CEL to a crystalline state owing to the presence of nuclei. Investigation of the residual solids revealed that supersaturation could be maintained for a while after the appearance of the crystals, which could be explained by heterogeneous nucleation and competition between the dissolution of amorphous parts and crystallization. In addition, a new crystalline form of CEL was observed during dissolution.
Collapse
Affiliation(s)
- Jingwen Song
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Ibaraki, Japan
| | - Kohsaku Kawakami
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Ibaraki, Japan
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Ibaraki, Japan
| |
Collapse
|
15
|
Tanaka H, Miyano T, Ueda H. Dissolution-permeation of hot-melt extruded amorphous solid dispersion comprising an experimental grade of HPMCAS. ADMET AND DMPK 2023; 11:373-385. [PMID: 37829323 PMCID: PMC10567069 DOI: 10.5599/admet.1586] [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: 11/04/2022] [Revised: 07/20/2023] [Indexed: 10/14/2023] Open
Abstract
Background and purpose Physicochemical properties of an amorphous solid dispersion (ASD) comprising an experimental grade of hydroxypropyl methylcellulose acetate succinate (HPMCAS-MX) with lower glass transition temperature have been previously investigated. This study aimed to evaluate applicability of HPMCAS-MX to hot-melt extrusion (HME) and dissolution-permeation performance of prepared ASDs using MicroFLUX. Review approach A physical mixture of indomethacin (IMC) and HPMCAS-MX or -MG (a commercial grade with higher transition temperature) at 20:80 weight ratio was hot-melt extruded to prepare an ASD (IMC-MX and IMC-MG, respectively). The dissolution-permeation performance and the stability of the ASDs were measured. Key results A torque reduction at 120 °C implied that IMC-MX transformed into an amorphous state at this temperature, but IMC-MG required around 170 °C. This result was supported by Raman mapping of the the HME samples. IMC-MG and IMC-MX remained in an amorphous state at 40 °C for three months. The initial dissolution rate and solubility of the ASDs were higher than that of crystalline IMC. The apparent permeability of IMC from IMC-MX and IMC-MG was comparable but was approximately two-fold higher than that from crystalline IMC. Conclusion HPMCAS-MX enabled HME process at a lower temperature and improved the dissolution-permeation performance of indomethacin.
Collapse
Affiliation(s)
- Hironori Tanaka
- Formulation R&D Laboratory, Shionogi & Co., Ltd., Hyogo 660-0813, Japan
| | - Tetsuya Miyano
- Laboratory for Medicinal Chemistry Research, Shionogi & Co., Ltd., Osaka, 561-0825, Japan
| | - Hiroshi Ueda
- Analysis and Evaluation Laboratory, Shionogi & Co., Ltd., Osaka, 561-0825, Japan
| |
Collapse
|
16
|
Kawakami K. Rigid Nuclei and Flexible Nuclei: Appearance and Disappearance of Nuclei in Indomethacin Glass Revealed by a Long-Term Annealing Study. J Phys Chem B 2023; 127:5967-5977. [PMID: 37408472 DOI: 10.1021/acs.jpcb.3c02127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
In this study, nucleation/crystallization behaviors of indomethacin glass are discussed with emphasis on the fate of nuclei, which is explained using a notion of "rigid" and "flexible" nuclei. The observation was made mainly by thermal analysis of indomethacin glass after long-term annealing at various temperatures. Formation of nuclei was evaluated by observing cold crystallization behaviors of the annealed glasses, as they should be dominated by the "nuclei form" produced in the glass. Nuclei of forms α and γ, which have opposite stability trends, were found to appear over a wide temperature range. The nuclei of form α were stable even in the presence of other crystal forms, whereas those of form γ were likely to be easily integrated into other crystals during their growth, which was explained by a notion of rigid and flexible nuclei. Moreover, unconventionally fast crystallization in the glass-transition region and the finding of a new crystal form are also reported.
Collapse
Affiliation(s)
- Kohsaku Kawakami
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| |
Collapse
|
17
|
Jiang J, Ouyang D, Williams RO. Predicting Glass-Forming Ability of Pharmaceutical Compounds by Using Machine Learning Technologies. AAPS PharmSciTech 2023; 24:103. [PMID: 37072563 DOI: 10.1208/s12249-023-02535-6] [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: 11/30/2022] [Accepted: 02/14/2023] [Indexed: 04/20/2023] Open
Abstract
Low aqueous solubility is a common and serious challenge for most drug substances not only in development but also in the market, and it may cause low absorption and bioavailability as a result. Amorphization is an intermolecular modification strategy to address the issue by breaking the crystal lattice and enhancing the energy state. However, due to the physicochemical properties of the amorphous state, drugs are thermodynamically unstable and tend to recrystallize over time. Glass-forming ability (GFA) is an experimental method to evaluate the forming and stability of glass formed by crystallization tendency. Machine learning (ML) is an emerging technique widely applied in pharmaceutical sciences. In this study, we successfully developed multiple ML models (i.e., random forest (RF), XGBoost, and support vector machine (SVM)) to predict GFA from 171 drug molecules. Two different molecular representation methods (i.e., 2D descriptor and Extended-connectivity fingerprints (ECFP)) were implemented to process the drug molecules. Among all ML algorithms, 2D-RF performed best with the highest accuracy, AUC, and F1 of 0.857, 0.850, and 0.828, respectively, in the testing set. In addition, we conducted a feature importance analysis, and the results mostly agreed with the literature, which demonstrated the interpretability of the model. Most importantly, our study showed great potential for developing amorphous drugs by in silico screening of stable glass formers.
Collapse
Affiliation(s)
- Junhuang Jiang
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, Texas, 78712, USA
| | - Defang Ouyang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, Macau, 999078, China
| | - Robert O Williams
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, Texas, 78712, USA.
| |
Collapse
|
18
|
Zemánková A, Hassouna F, Klajmon M, Fulem M. Solid–Liquid Equilibrium in Co-Amorphous Systems: Experiment and Prediction. Molecules 2023; 28:molecules28062492. [PMID: 36985463 PMCID: PMC10052153 DOI: 10.3390/molecules28062492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
In this work, the solid–liquid equilibrium (SLE) of four binary systems combining two active pharmaceutical ingredients (APIs) capable of forming co-amorphous systems (CAMs) was investigated. The binary systems studied were naproxen-indomethacin, naproxen-ibuprofen, naproxen-probucol, and indomethacin-paracetamol. The SLE was experimentally determined by differential scanning calorimetry. The thermograms obtained revealed that all binary mixtures investigated form eutectic systems. Melting of the initial binary crystalline mixtures and subsequent quenching lead to the formation of CAM for all binary systems and most of the compositions studied. The experimentally obtained liquidus and eutectic temperatures were compared to theoretical predictions using the perturbed-chain statistical associating fluid theory (PC-SAFT) equation of state and conductor-like screening model for real solvents (COSMO-RS), as implemented in the Amsterdam Modeling Suite (COSMO-RS-AMS). On the basis of the obtained results, the ability of these models to predict the phase diagrams for the investigated API–API binary systems was evaluated. Furthermore, the glass transition temperature (Tg) of naproxen (NAP), a compound with a high tendency to recrystallize, whose literature values are considerably scattered, was newly determined by measuring and modeling the Tg values of binary mixtures in which amorphous NAP was stabilized. Based on this analysis, erroneous literature values were identified.
Collapse
Affiliation(s)
- Alžběta Zemánková
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague, Czech Republic; (A.Z.); (M.K.)
| | - Fatima Hassouna
- Faculty of Chemical Engineering, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague, Czech Republic
| | - Martin Klajmon
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague, Czech Republic; (A.Z.); (M.K.)
| | - Michal Fulem
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague, Czech Republic; (A.Z.); (M.K.)
- Correspondence:
| |
Collapse
|
19
|
Indomethacin: Effect of Diffusionless Crystal Growth on Thermal Stability during Long-Term Storage. Molecules 2023; 28:molecules28041568. [PMID: 36838556 PMCID: PMC9963031 DOI: 10.3390/molecules28041568] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Differential scanning calorimetry and Raman spectroscopy were used to study the nonisothermal and isothermal crystallization behavior of amorphous indomethacin powders (with particle sizes ranging from 50 to 1000 µm) and their dependence on long-term storage conditions, either 0-100 days stored freely at laboratory ambient temperatures and humidity or placed in a desiccator at 10 °C. Whereas the γ-form polymorph always dominated, the accelerated formation of the α-form was observed in situations of heightened mobility (higher temperature and heating rate), increased amounts of mechanically induced defects, and prolonged free-surface nucleation. A complex crystallization behavior with two separated crystal growth modes (originating from either the mechanical defects or the free surface) was identified both isothermally and nonisothermally. The diffusionless glass-crystal (GC) crystal growth was found to proceed during the long-term storage at 10 °C and zero humidity, at the rate of ~100 µm of the γ-form surface crystalline layer being formed in 100 days. Storage at the laboratory temperature (still below the glass transition temperature) and humidity led only to a negligible/nondetectable GC growth for the fine indomethacin powders (particle size below ~150 µm), indicating a marked suppression of GC growth by the high density of mechanical defects under these conditions. The freely stored bulk material with no mechanical damage and a smooth surface exhibited zero traces of GC growth (as confirmed by microscopy) after >150 days of storage. The accuracy of the kinetic predictions of the indomethacin crystallization behavior was rather poor due to the combined influences of the mechanical defects, competing nucleation, and crystal growth processes of the two polymorphic phases as well as the GC growth complex dependence on the storage conditions within the vicinity of the glass transition temperature. Performing paired isothermal and nonisothermal kinetic measurements is thus highly recommended in macroscopic crystallization studies of drugs with similarly complicated crystal growth behaviors.
Collapse
|
20
|
Heczko D, Tarnacka M, Kamiński K, Paluch M, Kamińska E. Breakdown of isochronal superpositioning of α- and β-relaxation times in the van der Waals system – loratadine. J Mol Liq 2023. [DOI: 10.1016/j.molliq.2023.121377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
21
|
Wu H, Wang Z, Zhao Y, Gao Y, Zhang H, Wang L, Wang Z, Han J. Effect of Span 20 Feeding Zone in the Twin Screw Extruder on the Properties of Amorphous Solid Dispersion of Ritonavir. Pharmaceutics 2023; 15:pharmaceutics15020441. [PMID: 36839764 PMCID: PMC9960583 DOI: 10.3390/pharmaceutics15020441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/18/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
A ternary amorphous solid dispersion (ASD) system consisting of drug/polymer/surfactant is receiving increased attention to improve the oral bioavailability of poorly water-soluble drugs. The effect of polymers has been extensively studied, while the impact of surfactants has not yet to be studied to the same extent. Challenging questions to be answered are whether the surfactants should be added with the drug or separately and the resulting differences between the two operating processes. By adjusting the liquid feeding zone for Span 20 in the hot-melt twin screw extruder equipment, we investigated the effect of Span 20 on the properties of the polyvinylpyrrolidone/vinyl acetate (PVPVA)-based ASD formulations of ritonavir. We found that with the delayed feeding positions of Span 20 in the twin screw extruder, the ability of the ternary ASDs to maintain the supersaturation of the milled extrudates was observed to be significantly enhanced. Furthermore, adding surfactant after a thorough mixing of polymer and drug could decrease the molecular mobility of ternary ASD formulations. In addition, the effects of Span 20 on the complex viscosity and structure of PVPVA were also investigated. The delayed addition of Span 20 could improve the complex viscosity of PVPVA, thus leading to the drug precipitation inhibition. In conclusion, the delayed addition of Span 20 in the twin screw extruder and prolonging the mixing time of the drug and polymer may be critical to the maintenance of supersaturation.
Collapse
Affiliation(s)
- Hengqian Wu
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
- Institute of BioPharmaceutical Research, Liaocheng University, Liaocheng 252000, China
| | - Zhengping Wang
- Institute of BioPharmaceutical Research, Liaocheng University, Liaocheng 252000, China
- Liaocheng High-Tech Biotechnology Co., Ltd., Liaocheng 252059, China
| | - Yanna Zhao
- Institute of BioPharmaceutical Research, Liaocheng University, Liaocheng 252000, China
| | - Yan Gao
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Heng Zhang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Lili Wang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Zhe Wang
- Anhui Biochem Biopharmaceutical Co., Ltd., Hefei 230088, China
| | - Jun Han
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
- Institute of BioPharmaceutical Research, Liaocheng University, Liaocheng 252000, China
- Liaocheng High-Tech Biotechnology Co., Ltd., Liaocheng 252059, China
- Correspondence: ; Tel.: +86-0635-8239136
| |
Collapse
|
22
|
Shikha S, Lee YW, Doyle PS, Khan SA. Microfluidic Particle Engineering of Hydrophobic Drug with Eudragit E100─Bridging the Amorphous and Crystalline Gap. Mol Pharm 2022; 19:4345-4356. [PMID: 36268657 DOI: 10.1021/acs.molpharmaceut.2c00714] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Co-processing active pharmaceutical ingredients (APIs) with excipients is a promising particle engineering technique to improve the API physical properties, which can lead to more robust downstream drug product manufacturing and improved drug product attributes. Excipients provide control over critical API attributes like particle size and solid-state outcomes. Eudragit E100 is a widely used polymeric excipient to modulate drug release. Being cationic, it is primarily employed as a precipitation inhibitor to stabilize amorphous solid dispersions. In this work, we demonstrate how co-processing of E100 with naproxen (NPX) (a model hydrophobic API) into monodisperse emulsions via droplet microfluidics followed by solidification via solvent evaporation allows the facile fabrication of compact, monodisperse, and spherical particles with an expanded range of solid-state outcomes spanning from amorphous to crystalline forms. Low E100 concentrations (≤26% w/w) yield crystalline microparticles with a stable NPX polymorph distributed uniformly across the matrix at a high drug loading (∼89% w/w). Structurally, E100 incorporation reduces the size of primary particles comprising the co-processed microparticles in comparison to neat API microparticles made using the same technique and the as-received API powder. This reduction in primary particle size translates into an increased internal porosity of the co-processed microparticles, with specific surface area and pore volume ∼9 times higher than the neat API microparticles. These E100-enabled structural modifications result in faster drug release in acidic media compared to neat API microparticles. Additionally, E100-NPX microparticles have a significantly improved flowability compared to neat API microparticles and as-received API powder. Overall, this study demonstrates a facile microfluidics-based co-processing method that broadly expands the range of solid-state outcomes obtainable with E100 as an excipient, with multiscale control over the key attributes and performance of hydrophobic API-laden microparticles.
Collapse
Affiliation(s)
- Swati Shikha
- Critical Analytics for Manufacturing Personalized-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore138602, Singapore
| | - Yi Wei Lee
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore117576, Singapore.,NUS Graduate School for Integrative Sciences & Engineering, National University of Singapore, Singapore119077, Singapore
| | - Patrick S Doyle
- Critical Analytics for Manufacturing Personalized-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore138602, Singapore.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts02139, United States.,Harvard Medical School Initiative for RNA Medicine, Boston, Massachusetts02215, United States
| | - Saif A Khan
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore117576, Singapore
| |
Collapse
|
23
|
Svoboda R, Košťálová D, Krbal M, Komersová A. Indomethacin: The Interplay between Structural Relaxation, Viscous Flow and Crystal Growth. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27175668. [PMID: 36080433 PMCID: PMC9458118 DOI: 10.3390/molecules27175668] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022]
Abstract
Non-isothermal differential scanning calorimetry (DSC) was used to study the influences of particle size (daver) and heating rate (q+) on the structural relaxation, crystal growth and decomposition kinetics of amorphous indomethacin. The structural relaxation and decomposition processes exhibited daver-independent kinetics, with the q+ dependences based on the apparent activation energies of 342 and 106 kJ·mol-1, respectively. The DSC-measured crystal growth kinetics played a dominant role in the nucleation throughout the total macroscopic amorphous-to-crystalline transformation: the change from the zero-order to the autocatalytic mechanism with increasing q+, the significant alteration of kinetics, with the storage below the glass transition temperature, and the accelerated crystallization due to mechanically induced defects. Whereas slow q+ led to the formation of the thermodynamically stable γ polymorph, fast q+ produced a significant amount of the metastable α polymorph. Mutual correlations between the macroscopic and microscopic crystal growth processes, and between the viscous flow and structural relaxation motions, were discussed based on the values of the corresponding activation energies. Notably, this approach helped us to distinguish between particular crystal growth modes in the case of the powdered indomethacin materials. Ediger's decoupling parameter was used to quantify the relationship between the viscosity and crystal growth. The link between the cooperativity of structural domains, parameters of the Tool-Narayanaswamy-Moynihan relaxation model and microscopic crystal growth was proposed.
Collapse
Affiliation(s)
- Roman Svoboda
- Department of Physical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 532 10 Pardubice, Czech Republic
- Correspondence: ; Tel.: +420-466-037-420
| | - Daniela Košťálová
- Department of Physical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 532 10 Pardubice, Czech Republic
| | - Miloš Krbal
- Center of Materials and Nanotechnologies (CEMNAT), Faculty of Chemical Technology, University of Pardubice, nam. Cs legii 565, 530 02 Pardubice, Czech Republic
| | - Alena Komersová
- Department of Physical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 532 10 Pardubice, Czech Republic
| |
Collapse
|
24
|
Bookwala M, Buckner IS, Wildfong PLD. Implications of Coexistent Halogen and Hydrogen Bonds in Amorphous Solid Dispersions on Drug Solubility, Miscibility, and Mobility. Mol Pharm 2022; 19:3959-3972. [PMID: 36049226 DOI: 10.1021/acs.molpharmaceut.2c00434] [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: 11/28/2022]
Abstract
Specific noncovalent drug-polymer interactions were analytically identified using Raman and Fourier transform infrared spectroscopy for amorphous solid dispersions (ASD) formed between either chlorpropamide or tolbutamide and polyvinylpyrrolidone vinyl acetate random copolymer (PVPVA). Spectral changes in the C-Cl stretching vibrations due to changes in the electronic environment of the Cl atom confirmed halogen bond formation in chlorpropamide-PVPVA ASDs, the extent of which was established to be inversely related to the concentration of the drug using 2D correlation spectroscopy analysis. Hydrogen bonding between the secondary amide of each drug and the pyrrolidone carbonyl of the copolymer was also confirmed in all dispersions. Implications of coexistent interactions were investigated for drug-polymer solubility, mixing free energy, and molecular mobility relative to tolbutamide, which only formed hydrogen bonds with PVPVA. Chlorpropamide had a higher solubility, a larger negative mixing free energy, and lower mobility in PVPVA relative to tolbutamide. These thermodynamic and kinetic differences demonstrate the significance of halogen bond formation even when hydrogen bonding is present.
Collapse
Affiliation(s)
- Mustafa Bookwala
- School of Pharmacy and Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Ave, Pittsburgh, Pennsylvania 15282, United States
| | - Ira S Buckner
- School of Pharmacy and Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Ave, Pittsburgh, Pennsylvania 15282, United States
| | - Peter L D Wildfong
- School of Pharmacy and Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Ave, Pittsburgh, Pennsylvania 15282, United States
| |
Collapse
|
25
|
Study of Thermal Properties, Molecular Dynamics, and Physical Stability of Etoricoxib Mixtures with Octaacetylmaltose near the Glass Transition. Int J Mol Sci 2022; 23:ijms23179794. [PMID: 36077212 PMCID: PMC9456116 DOI: 10.3390/ijms23179794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
In this paper, we thoroughly investigated the physical stability of the anti-inflammatory drug etoricoxib, which has been reported earlier to be resistant to recrystallization in its glassy and supercooled states at ambient pressure. Our unique application of the standard refractometry technique showed that the supercooled liquid of the drug was able to recrystallize during isothermal experiments in atmospheric conditions. This enabled us to determine the crystallization onset timescale and nucleation energy barrier of etoricoxib for the first time. As the physical instability of etoricoxib requires working out an efficient method for improving the drug’s resistance to recrystallization to maintain its amorphous form utility in potential pharmaceutical applications, we focused on finding a solution to this problem, and successfully achieved this purpose by preparing binary mixtures of etoricoxib with octaacetylmaltose. Our detailed thermal, refractometry, and molecular dynamics studies of the binary compositions near the glass transition revealed a peculiar behavior of the glass transition temperatures when changing the acetylated disaccharide concentration in the mixtures. Consequently, the anti-plasticization effect on the enhancement of physical stability could be excluded, and a key role for specific interactions in the improved resistance to recrystallization was expected. Invoking our previous results obtained for etoricoxib, the chemically similar drug celecoxib, and octaacetylmaltose, we formulated a hypothesis about the molecular mechanisms that may cause an impediment to crystal nuclei formation in the amorphous mixtures of etoricoxib with octaacetylmaltose. The most plausible scenario may rely on the formation of hydrogen-bonded heterodimers of the drug and excipient molecules, and the related drop in the population of the etoricoxib homodimers, which disables the nucleation. Nevertheless, this hypothesis requires further investigation. Additionally, we tested some widely discussed correlations between molecular mobility and crystallization properties, which turned out to be only partially satisfied for the examined mixtures. Our findings constitute not only a warning against manufacturing the amorphous form of pure etoricoxib, but also evidence for a promising outcome for the pharmaceutical application of the amorphous compositions with octaacetylmaltose.
Collapse
|
26
|
Flynn JJ, Marsh ZM, Krein DM, Wolf SM, Haley JE, Vasquez ES, Cooper TM, Godman NP, Grusenmeyer TA. Identification of Lithocholic Acid as a Molecular Glass Host for Room‐Temperature Phosphorescent Materials. CHEMPHOTOCHEM 2022. [DOI: 10.1002/cptc.202200134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- John J. Flynn
- Air Force Research Laboratory Materials & Manufacturing Directorate Photonic Materials Branch UNITED STATES
| | - Zachary M. Marsh
- Air Force Research Laboratory Materials & Manufacturing Directorate Photonic Materials Branch UNITED STATES
| | - Douglas M. Krein
- Air Force Research Laboratory Materials & Manufacturing Directorate Photonic Materials Branch UNITED STATES
| | - Steven M. Wolf
- Air Force Research Laboratory Materials & Manufacturing Directorate Photonic Materials Branch UNITED STATES
| | - Joy E. Haley
- Air Force Research Laboratory Materials & Manufacturing Directorate Photonic Materials Branch UNITED STATES
| | - Erick S. Vasquez
- University of Dayton Department of Chemical and Materials Engineering UNITED STATES
| | - Thomas M. Cooper
- Air Force Research Laboratory Materials & Manufacturing Directorate Photonic Materials Branch UNITED STATES
| | - Nicholas P. Godman
- Air Force Research Laboratory Materials & Manufacturing Directorate Photonic Materials Branch UNITED STATES
| | - Tod A. Grusenmeyer
- Air Force Research Laboratory Materials & Manufacturing Directorate Photonic Materials Branch 2179 12th Street Bldg 652 45433 Wright-Patterson AFB UNITED STATES
| |
Collapse
|
27
|
A Comparative Study of the Pharmaceutical Properties between Amorphous Drugs Loaded-Mesoporous Silica and Pure Amorphous Drugs Prepared by Solvent Evaporation. Pharmaceuticals (Basel) 2022; 15:ph15060730. [PMID: 35745649 PMCID: PMC9228546 DOI: 10.3390/ph15060730] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/27/2022] [Accepted: 06/02/2022] [Indexed: 12/05/2022] Open
Abstract
The formulation of poorly water-soluble drugs is one of the main challenges in the pharmaceutical industry, especially in the development of oral dosage forms. Meanwhile, there is an increase in the number of poorly soluble drugs that have been discovered as new chemical entities. It was also reported that the physical transformation of a drug from a crystalline form into an amorphous state could be used to increase its solubility. Therefore, this study aims to evaluate the pharmaceutical properties of amorphous drug loaded-mesoporous silica (MPS) and pure amorphous drugs. Ritonavir (RTV) was used as a model of a poorly water-soluble drug due to its low recrystallization tendency. RTV loaded-MPS (RTV/MPS) and RTV amorphous were prepared using the solvent evaporation method. Based on observation, a halo pattern in the powder X-ray diffraction pattern and a single glass transition (Tg) in the modulated differential scanning calorimetry (MDSC) curve was discovered in RTV amorphous, indicating its amorphization. The Tg was not detected in RTV/MPS, which showed that the loading RTV was completed. The solid-state NMR and FT-IR spectroscopy also showed the interaction between RTV and the surface of MPS in the mesopores. The high supersaturation of RTV was not achieved for both RTV/MPS and the amorphous state due to its strong interaction with the surface of MPS and was not properly dispersed in the medium, respectively. In the dissolution test, the molecular dispersion of RTV within MPS caused rapid dissolution at the beginning, while the amorphous showed a low rate due to its agglomeration. The stability examination showed that the loading process significantly improved the physical and chemical stability of RTV amorphous. These results indicated that the pharmaceutical properties of amorphous drugs could be improved by loaded-MPS.
Collapse
|
28
|
Amorphous characterization of pharmaceutical drug substances enabled by the elastic modulus mapping of atomic force microscope. Int J Pharm 2022; 621:121784. [DOI: 10.1016/j.ijpharm.2022.121784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 11/19/2022]
|
29
|
Ueda H, Hirakawa Y, Miyano T, Imono M, Tse JY, Uchiyama H, Tozuka Y, Kadota K. Design of a Stable Coamorphous System Using Lactose as an Antiplasticizing Agent for Diphenhydramine Hydrochloride with a Low Glass Transition Temperature. Mol Pharm 2022; 19:1209-1218. [PMID: 35316068 DOI: 10.1021/acs.molpharmaceut.2c00057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Coamorphous systems comprising small molecules are emerging as counterparts to polymeric solid dispersions. However, the glass transition temperatures (Tgs) of coamorphous materials are relatively low because of the lack of polymeric carriers with higher Tgs. This study aimed to investigate the applicability of lactose (LAC) as an antiplasticizing coformer to a coamorphous system. Diphenhydramine hydrochloride (DPH) was selected as a model drug (Tg = 16 °C). Differential scanning calorimetry showed a comelting point in addition to a decrease in the neat melting points depending on the composition of the physical mixtures, suggesting that the mixture of DPH-LAC was eutectic. The melting point of the eutectic mixture was calculated according to the Schröder-van Laar equation. The heat of fusion of the eutectic mixture was maximized at a 70:30 molar ratio of DPH to LAC; at this point, the melting peaks of the pure components disappeared. The heat flow profiles following the melting and cooling of DPH-LAC physical mixtures at the ratios from 10:90 to 90:10 showed a single Tg, suggesting the formation of a coamorphous system. Lactose showed a Tg of over 100 °C, and the Tg of DPH increased with the molar ratio of LAC; it was 84 °C at a 10:90 molar ratio of DPH to LAC. The Raman image indicated the formation of a homogeneous dispersion of DPH and LAC in the coamorphous system. Peak shifts in the infrared spectra indicated the presence of intermolecular interactions between the amino group of DPH and the hydroxyl group of LAC. Principal component analysis of the infrared spectra revealed a significant change at the 70:30 molar ratio of DPH to LAC, which was in agreement with the results of the thermal analysis. A stability test at 40 °C revealed rapid crystallization of the supercooled liquid DPH. The coamorphous samples containing 10-50% of LAC remained in an amorphous state for 21 days, and no crystallization was observed for the samples containing >60% of LAC for 28 days. The relatively lower Tg (less than 40 °C) of the coamorphous system containing 10-50% of LAC might have caused crystallization during storage. These findings indicate that LAC, which is a safe and widely used pharmaceutical excipient, can be applied to coamorphous systems as an antiplasticizing coformer.
Collapse
Affiliation(s)
- Hiroshi Ueda
- Physical Chemistry, Laboratory for Medicinal Chemistry Research, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Yuya Hirakawa
- Chemical Modality 2, Laboratory for Advanced Medicine Research, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Tetsuya Miyano
- Physical Chemistry, Laboratory for Medicinal Chemistry Research, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Masaaki Imono
- Physical Chemistry, Laboratory for Medicinal Chemistry Research, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Jun Yee Tse
- Department of Formulation Design and Pharmaceutical Technology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka 569-1094, Japan
| | - Hiromasa Uchiyama
- Department of Formulation Design and Pharmaceutical Technology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka 569-1094, Japan
| | - Yuichi Tozuka
- Department of Formulation Design and Pharmaceutical Technology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka 569-1094, Japan
| | - Kazunori Kadota
- Department of Formulation Design and Pharmaceutical Technology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka 569-1094, Japan
| |
Collapse
|
30
|
Govender R, Abrahmsén-Alami S, Folestad S, Olsson M, Larsson A. Enabling modular dosage form concepts for individualized multidrug therapy: Expanding the design window for poorly water-soluble drugs. Int J Pharm 2021; 602:120625. [PMID: 33892062 DOI: 10.1016/j.ijpharm.2021.120625] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 12/13/2022]
Abstract
Multidrug dosage forms (aka combination dosage forms, polypills, etc.) create value for patients through reduced pill burdens and simplified administration to improve adherence to therapy. Enhanced flexibility of multidrug dosage forms would provide further opportunities to better match emerging needs for individualized therapy. Through modular dosage form concepts, one approach to satisfy these needs is to adapt multidrug dosage forms to a wider variety of drugs, each with a variety of doses and release profiles. This study investigates and technically explores design requirements for extending the capability of modular multidrug dosage form concepts towards individualization. This builds on our recent demonstration of independent tailoring of dose and drug release, which is here extended towards poorly water-soluble drugs. The challenging design requirement of carrying higher drug loads in smaller volumes to accommodate multiple drugs at their clinical dose is here met regarding dose and release performance. With a modular concept, we demonstrate high precision (<5% RSD) in dose and release performance of individual modules containing felodipine or naproxen in Kollidon VA64 at both a wide drug loading range (5% w/w and 50% w/w drug) and a small module size (3.6 mg). In a forward-looking design-based discussion, further requirements are addressed, emphasizing that reproducible individual module performance is predictive of dosage form performance, provided the modules are designed to act independently. Therefore, efforts to incorporate progressively higher drug loads within progressively smaller module volumes will be crucial to extend the design window further towards full flexibility of future dosage forms for individualized multidrug therapy.
Collapse
Affiliation(s)
- Rydvikha Govender
- Oral Product Development, Pharmaceutical Technology and Development, Operations, AstraZeneca, SE-43183 Gothenburg, Sweden; Pharmaceutical Technology, Chemistry and Chemical Engineering, Chalmers University of Technology, SE-41296 Gothenburg, Sweden.
| | - Susanna Abrahmsén-Alami
- Oral Product Development, Pharmaceutical Technology and Development, Operations, AstraZeneca, SE-43183 Gothenburg, Sweden
| | - Staffan Folestad
- Innovation Strategies and External Liaison, Pharmaceutical Technology and Development, Operations, AstraZeneca, SE-43183 Gothenburg, Sweden
| | - Martina Olsson
- Department of Physics, Chalmers University of Technology, SE-41296 Gothenburg, Sweden
| | - Anette Larsson
- Pharmaceutical Technology, Chemistry and Chemical Engineering, Chalmers University of Technology, SE-41296 Gothenburg, Sweden
| |
Collapse
|
31
|
Ho LY, Xiang ZS, Gopal R, Khan SA. Microfluidics-enabled particle engineering of monodisperse solid lipid microparticles with uniform drug loading and diverse solid-state outcomes. Int J Pharm 2021; 596:120230. [PMID: 33484918 DOI: 10.1016/j.ijpharm.2021.120230] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/23/2020] [Accepted: 12/19/2020] [Indexed: 11/19/2022]
Abstract
Lipids serve as excellent excipients for drug products. Solid lipid microparticles (SLMs) are relatively underexplored in drug delivery; these particles are conventionally prepared using processes yielding polydisperse size distributions, such as spray congealing or emulsification. In this paper, we demonstrate a microfluidics-enabled process for particle engineering of monodisperse solid lipid microparticles with size and content uniformity. To overcome low solubility, we use a volatile solvent to increase drug loading, making the drug-lipid solution a single phase, enabling identical drug loading across particles. We use microfluidic flow extrusion of the solution to generate uniform drug-loaded SLMs, substantially enhancing monodispersity. This method generalises across three drugs-ibuprofen, 5-methyl-2-[(2-nitrophenyl)amino]-3-thiophenecarbonitrile (ROY), and naproxen, and two lipids-beeswax and hard fat (Suppocire NAI 25A), forming particles of various solid states: amorphous naproxen in crystalline lipids, crystalline ROY in crystalline lipids, and a eutectic mixture of ibuprofen-hard fat. In vitro dissolution studies on the ibuprofen-hard fat SLMs reveal gradual release, fitting the Higuchi model with 50-65% drug released over 72 h. This work expands the drug particle engineering toolbox to enable the formulation of SLMs with high precision in particle size and drug loading. Moreover, the diverse solid-state outcomes enabled by our method makes it applicable to various drugs having different formulation requirements (crystalline/amorphous).
Collapse
Affiliation(s)
- Leon Yoon Ho
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117576, Singapore
| | - Zi Shun Xiang
- Matralix Pte Ltd, 71 Ayer Rajah Crescent, #07-05, Singapore 139951, Singapore
| | - Renuga Gopal
- Matralix Pte Ltd, 71 Ayer Rajah Crescent, #07-05, Singapore 139951, Singapore
| | - Saif A Khan
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117576, Singapore.
| |
Collapse
|
32
|
Jelić D. Thermal Stability of Amorphous Solid Dispersions. Molecules 2021; 26:E238. [PMID: 33466393 PMCID: PMC7795217 DOI: 10.3390/molecules26010238] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/25/2020] [Accepted: 12/29/2020] [Indexed: 11/24/2022] Open
Abstract
Amorphous solid dispersion drug delivery systems (ASD DDS) were proved to be efficient for the enhancement of solubility and bioavailability of poorly water-soluble drugs. One of the major keys for successful preparation of ASD is the selection of appropriate excipients, mostly polymers, which have a crucial role in improving drug solubility and its physical stability. Even though, excipients should be chemically inert, there is some evidence that polymers can affect the thermal stability of active pharmaceutical ingredients (API). The thermal stability of a drug is closely related to the shelf-life of pharmaceutical products and therefore it is a matter of high pharmaceutical relevance. An overview of thermal stability of amorphous solids is provided in this paper. Evaluation of thermal stability of amorphous solid dispersion is perceived from the physicochemical perspective, from a kinetic (motions) and thermodynamic (energy) point of view, focusing on activation energy and fragility, as well all other relevant parameters for ASD design, with a glance on computational kinetic analysis of solid-state decomposition.
Collapse
Affiliation(s)
- Dijana Jelić
- Chemistry Department, Faculty of Natural Sciences and Mathematics, University of Banja Luka, dr Mladena Stojanovića 2a, 78 000 Banja Luka, Bosnia and Herzegovina
| |
Collapse
|
33
|
Bioavailability Improvement of Carbamazepine via Oral Administration of Modified-Release Amorphous Solid Dispersions in Rats. Pharmaceutics 2020; 12:pharmaceutics12111023. [PMID: 33114739 PMCID: PMC7693946 DOI: 10.3390/pharmaceutics12111023] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 11/17/2022] Open
Abstract
The purpose of this study was to improve the bioavailability of carbamazepine (CBZ), a poorly water-soluble antiepileptic drug, via modified-release amorphous solid dispersions (mr-ASD) by a thin film freezing (TFF) process. Three types of CBZ-mr-ASD with immediate-, delayed-, and controlled-release properties were successfully prepared with HPMC E3 (hydrophilic), L100-55 (enteric), and cellulose acetate (CA, lipophilic), defined as CBZ-ir-ASD, CBZ-dr-ASD, and CBZ-cr-ASD, respectively. A dry granulation method was used to prepare CBZ-mr-ASD capsule formulations. Various characterization techniques were applied to evaluate the physicochemical properties of CBZ-mr-ASD and the related capsules. The drug remained in an amorphous state when encapsulated within CBZ-mr-ASD, and the capsule formulation progress did not affect the performance of the dispersions. In dissolution tests, the preparations and the corresponding dosage forms similarly showed typical immediate-, delayed-, and controlled-release properties depending on the solubility of the polymers. Moreover, single-dose 24 h pharmacokinetic studies in rats indicated that CBZ-mr-ASD significantly enhanced the oral absorption of CBZ compared to that of crude CBZ. Increased oral absorption of CBZ was observed, especially in the CBZ-dr-ASD formulation, which showed a better pharmacokinetic profile than that of crude CBZ with 2.63- and 3.17-fold improved bioavailability of the drug and its main active metabolite carbamazepine 10,11-epoxide (CBZ-E).
Collapse
|
34
|
Ditzinger F, Wieland R, Statelova M, Vertzoni M, Holm R, Kuentz M. In Vivo Performance of Innovative Polyelectrolyte Matrices for Hot Melt Extrusion of Amorphous Drug Systems. Mol Pharm 2020; 17:3053-3061. [PMID: 32589437 DOI: 10.1021/acs.molpharmaceut.0c00485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hot melt extrusion of amorphous systems has become a pivotal technology to cope with challenges of poorly water-soluble drugs. Previous research showed that small molecular additives with targeted molecular interactions enabled introduction of a polyelectrolyte matrix into hot melt extrusion that would otherwise not be possible to process due to the unfavorable properties upon heating of the pure polymer. Carboxymethyl cellulose sodium (NaCMC) with lysine or alternatively meglumine led to modified polymeric matrices that showed adequate processability by hot melt extrusion and yielded stable amorphous formulations. The investigated formulations, including fenofibrate as a model drug, were characterized by attenuated total reflectance Fourier transform infrared spectroscopy, differential scanning calorimetry, and viscosity measurements after aqueous dispersion. Further biopharmaceutical assessment started with biorelevant nonsink dissolution testing followed by a pharmacokinetic in vivo study in rats. The in vitro assessment showed superiority of the lysine-containing formulation in the extent of in vitro supersaturation and overall drug release. In accordance with this, the in vivo study also demonstrated increased exposure of the amorphous formulations and in particular for the system containing lysine. In summary, the combination of polyelectrolytes with interacting additives presents a promising opportunity for the formulation of poorly water-soluble drugs.
Collapse
Affiliation(s)
- Felix Ditzinger
- Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland.,Institute of Pharma Technology, University of Applied Sciences and Arts Northwestern Switzerland, 4132 Muttenz, Switzerland
| | - Rebecca Wieland
- Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland
| | - Marina Statelova
- Department of Pharmacy, National and Kapodistrian University of Athens, 157 84 Athens, Greece
| | - Maria Vertzoni
- Department of Pharmacy, National and Kapodistrian University of Athens, 157 84 Athens, Greece
| | - René Holm
- Drug Product Development, Janssen Research and Development, Johnson and Johnson, 2340 Beerse, Belgium.,Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark
| | - Martin Kuentz
- Institute of Pharma Technology, University of Applied Sciences and Arts Northwestern Switzerland, 4132 Muttenz, Switzerland
| |
Collapse
|
35
|
Blasi P, Casagrande S, Pedretti A, Fioretto D, Vistoli G, Corezzi S. Ketoprofen poly(lactide-co-glycolide) physical interaction studied by Brillouin spectroscopy and molecular dynamics simulations. Int J Pharm 2020; 580:119235. [DOI: 10.1016/j.ijpharm.2020.119235] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/09/2020] [Accepted: 03/14/2020] [Indexed: 12/20/2022]
|
36
|
Kawakami K, Suzuki K, Fukiage M, Matsuda M, Nishida Y, Oikawa M, Fujita T. Impact of degree of supersaturation on the dissolution and oral absorption behaviors of griseofulvin amorphous solid dispersions. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2019.101172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
37
|
Opportunities for Successful Stabilization of Poor Glass-Forming Drugs: A Stability-Based Comparison of Mesoporous Silica Versus Hot Melt Extrusion Technologies. Pharmaceutics 2019; 11:pharmaceutics11110577. [PMID: 31689980 PMCID: PMC6920921 DOI: 10.3390/pharmaceutics11110577] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/25/2019] [Accepted: 11/01/2019] [Indexed: 11/17/2022] Open
Abstract
Amorphous formulation technologies to improve oral absorption of poorly soluble active pharmaceutical ingredients (APIs) have become increasingly prevalent. Currently, polymer-based amorphous formulations manufactured by spray drying, hot melt extrusion (HME), or co-precipitation are most common. However, these technologies have challenges in terms of the successful stabilization of poor glass former compounds in the amorphous form. An alternative approach is mesoporous silica, which stabilizes APIs in non-crystalline form via molecular adsorption inside nano-scale pores. In line with these considerations, two poor glass formers, haloperidol and carbamazepine, were formulated as polymer-based solid dispersion via HME and with mesoporous silica, and their stability was compared under accelerated conditions. Changes were monitored over three months with respect to solid-state form and dissolution. The results were supported by solid-state nuclear magnetic resonance spectroscopy (SS-NMR) and scanning electron microscopy (SEM). It was demonstrated that mesoporous silica was more successful than HME in the stabilization of the selected poor glass formers. While both drugs remained non-crystalline during the study using mesoporous silica, polymer-based HME formulations showed recrystallization after one week. Thus, mesoporous silica represents an attractive technology to extend the formulation toolbox to poorly soluble poor glass formers.
Collapse
|
38
|
Abstract
Stabilization technology of glass structures is of great interest in the field of pharmaceutical science, as it may prevent poorly soluble candidate compounds from dropping out of the pipeline. Cooling rate from the melt has been recognized as one parameter to alter the energy state of the glass; however, the relationship between the physicochemical properties of glass and stabilization efficiency of the cooling rate has not been clarified yet. We have investigated the effect of cooling rate on the thermodynamic parameters of 13 pharmaceutical glasses, to find features of the compounds that are closely related to the stabilization efficiency. We have also analyzed the structural differences between slowly cooled and annealed glasses based on Fourier-transform infrared spectra and relaxation enthalpy. Although the degree of stabilization was lower for slowly cooled glasses compared to that for vapor-deposited ones, slow cooling was found to be a prominent method for producing stable glass and is applicable to bulk materials. In this observation, a strong correlation between fragility and the number of rotatable bonds was also found.
Collapse
Affiliation(s)
- Kohsaku Kawakami
- International Center for Materials Nanoarchitectonics , National Institute for Materials Science , 1-1 Namiki , Tsukuba , Ibaraki 305-0044 , Japan
| |
Collapse
|