1
|
Queiroz LHS, Barros RS, de Sousa FF, Lage MR, Sarraguça MC, Ribeiro PRS. Preparation and Characterization of a Rifampicin Coamorphous Material with Tromethamine Coformer: An Experimental-Theoretical Study. Mol Pharm 2024; 21:1272-1284. [PMID: 38361428 DOI: 10.1021/acs.molpharmaceut.3c00947] [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: 02/17/2024]
Abstract
Rifampicin (RIF) is an antibiotic used to treat tuberculosis and leprosy. Even though RIF is a market-available drug, it has a low aqueous solubility, hindering its bioavailability. Among the strategies for bioavailability improvement of poorly soluble drugs, coamorphous systems have been revealed as an alternative in the increase of the aqueous solubility of drug systems and at the same time also increasing the amorphous state stability and dissolution rate when compared with the neat drug. In this work, a new coamorphous form from RIF and tromethamine (TRIS) was synthesized by slow evaporation. Structural, electronic, and thermodynamic properties and solvation effects, as well as drug-coformer intermolecular interactions, were studied through density functional theory (DFT) calculations. Powder X-ray diffraction (PXRD) data allowed us to verify the formation of a new coamorphous. In addition, the DFT study indicates a possible intermolecular interaction by hydrogen bonds between the available amino and carbonyl groups of RIF and the hydroxyl and amino groups of TRIS. The theoretical spectra obtained are in good agreement with the experimental data, suggesting the main interactions occurring in the formation of the coamorphous system. PXRD was used to study the physical stability of the coamorphous system under accelerated ICH conditions (40 °C and 75% RH), indicating that the material remained in an amorphous state up to 180 days. The thermogravimetry result of this material showed a good thermal stability up to 153 °C, and differential scanning calorimetry showed that the glass temperature (Tg) was at 70.0 °C. Solubility studies demonstrated an increase in the solubility of RIF by 5.5-fold when compared with its crystalline counterpart. Therefore, this new material presents critical parameters that can be considered in the development of new coamorphous formulations.
Collapse
Affiliation(s)
- Luís H S Queiroz
- NUPFARQ, Programa de Pós-Graduação em Ciência dos Materiais (PPGCM), Centro de Ciências de Imperatriz (CCIM), Universidade Federal do Maranhão (UFMA), Imperatriz, Maranhão 65.900-410, Brazil
| | - Ranna S Barros
- NUPFARQ, Programa de Pós-Graduação em Ciência dos Materiais (PPGCM), Centro de Ciências de Imperatriz (CCIM), Universidade Federal do Maranhão (UFMA), Imperatriz, Maranhão 65.900-410, Brazil
| | - Francisco F de Sousa
- Instituto de Ciências Exatas e Naturais, Universidade Federal do Pará (UFPA), Belém, Pará 65.075-110, Brazil
| | - Mateus R Lage
- NUPFARQ, Programa de Pós-Graduação em Ciência dos Materiais (PPGCM), Centro de Ciências de Imperatriz (CCIM), Universidade Federal do Maranhão (UFMA), Imperatriz, Maranhão 65.900-410, Brazil
- Coordenação do Curso de Ciência e Tecnologia, Centro de Ciências de Balsas, Universidade Federal do Maranhão (UFMA), Balsas, Maranhão 65.800-000, Brazil
| | - Mafalda C Sarraguça
- LAQV, REQUIMTE, Department of Chemical Sciences, Laboratory of Applied Chemistry, Faculty of Pharmacy, Porto University, Rua de Jorge Viterbo Ferreira, 228, Porto 4050-313, Portugal
| | - Paulo R S Ribeiro
- NUPFARQ, Programa de Pós-Graduação em Ciência dos Materiais (PPGCM), Centro de Ciências de Imperatriz (CCIM), Universidade Federal do Maranhão (UFMA), Imperatriz, Maranhão 65.900-410, Brazil
| |
Collapse
|
2
|
Wang H, Zhao P, Ma R, Jia J, Fu Q. Drug-drug co-amorphous systems: An emerging formulation strategy for poorly water-soluble drugs. Drug Discov Today 2024; 29:103883. [PMID: 38219970 DOI: 10.1016/j.drudis.2024.103883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/21/2023] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
Overcoming the poor water solubility of small-molecule drugs is a major challenge in the development of clinical pharmaceuticals. Amorphization of crystalline drugs is a highly effective strategy to improve their aqueous solubility. However, amorphous drugs are thermodynamically unstable and likely to crystallize during manufacturing and storage. Recently, drug-drug co-amorphous systems have emerged as a novel strategy to not only enable enhanced dissolution and physical stability of the individual drugs within the system but also to provide a strategy for combination therapy of the same or different clinical indications. This review serves to highlight advances in the methods used to manufacture and characterize drug-drug co-amorphous systems, summarize drug-drug co-amorphous applications reported in recent decades, and provide an outlook on future possibilities and perspectives.
Collapse
Affiliation(s)
- Hongge Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Peixu Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Ruilong Ma
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Jirun Jia
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Qiang Fu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
3
|
Di R, Rades T, Grohganz H. Destabilization of Indomethacin-Paracetamol Co-Amorphous Systems by Mechanical Stress. Pharmaceutics 2023; 16:67. [PMID: 38258078 PMCID: PMC10818836 DOI: 10.3390/pharmaceutics16010067] [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: 11/17/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
Using co-amorphous systems (CAMS) has shown promise in addressing the challenges associated with poorly water-soluble drugs. Quench-cooling is a commonly used CAMS preparation method, often followed by grinding or milling to achieve a fine powder that is suitable for subsequent characterization or further down-stream manufacturing. However, the impact of mechanical stress applied to CAMS has received little attention. In this study, the influence of mechanical stress on indomethacin-paracetamol CAMS was investigated. The investigation involved thermal analysis and solid-state characterization across various CAMS mixing ratios and levels of mechanical stress. The study revealed a negative effect of mechanical stress on stability, particularly on the excess components in CAMS. Higher levels of mechanical stress were observed to induce phase separation or recrystallization. Notably, samples at the optimal mixing ratio demonstrated greater resistance to the destabilization caused by mechanical stress. These results showed the significance of careful consideration of processing methods during formulation and the significance of optimizing mixing ratios in CAMS.
Collapse
Affiliation(s)
| | - Thomas Rades
- Department of Pharmacy, University of Copenhagen, 2200 Copenhagen, Denmark; (R.D.); (H.G.)
| | | |
Collapse
|
4
|
Kapoor DU, Singh S, Sharma P, Prajapati BG. Amorphization of Low Soluble Drug with Amino Acids to Improve Its Therapeutic Efficacy: a State-of-Art-Review. AAPS PharmSciTech 2023; 24:253. [PMID: 38062314 DOI: 10.1208/s12249-023-02709-2] [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: 05/20/2023] [Accepted: 11/19/2023] [Indexed: 12/18/2023] Open
Abstract
Low aqueous solubility of drug candidates is an ongoing challenge and pharmaceutical manufacturers pay close attention to amorphization (AMORP) technology to improve the solubility of drugs that dissolve poorly. Amorphous drug typically exhibits much higher apparent solubility than their crystalline form due to high energy state that enable them to produce a supersaturated state in the gastrointestinal tract and thereby improve bioavailability. The stability and augmented solubility in co-amorphous (COA) formulations is influenced by molecular interactions. COA are excellent carriers-based drug delivery systems for biopharmaceutical classification system (BCS) class II and class IV drugs. The three important critical quality attributes, such as co-formability, physical stability, and dissolution performance, are necessary to illustrate the COA systems. New amorphous-stabilized carriers-based fabrication techniques that improve drug loading and degree of AMORP have been the focus of emerging AMORP technology. Numerous low-molecular-weight compounds, particularly amino acids such as glutamic acid, arginine, isoleucine, leucine, valine, alanine, glycine, etc., have been employed as potential co-formers. The review focus on the prevailing drug AMORP strategies used in pharmaceutical research, including in situ AMORP, COA systems, and mesoporous particle-based methods. Moreover, brief characterization techniques and the application of the different amino acids in stabilization and solubility improvements have been related.
Collapse
Affiliation(s)
| | - Sudarshan Singh
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Office of Research Administration, Faculty of Pharmacy, Chiang Mai University, 50200, Chiang Mai, Thailand.
| | - Pratishtha Sharma
- School of Pharmacy, Raffles University, Neemrana, Rajasthan, 301020, India
| | - Bhupendra G Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva, 384012, India.
| |
Collapse
|
5
|
Yusuf H, Meidy Nurintan Savitri O, Primaharinastiti R, Agus Syamsur Rijal M. A lyophilized-surfactant-based rutin formulation with improved physical characteristics and dissolution for oral delivery. Saudi Pharm J 2023. [DOI: 10.1016/j.jsps.2023.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023] Open
|
6
|
Shi Q, Wang Y, Moinuddin SM, Feng X, Ahsan F. Co-amorphous Drug Delivery Systems: a Review of Physical Stability, In Vitro and In Vivo Performance. AAPS PharmSciTech 2022; 23:259. [PMID: 36123515 DOI: 10.1208/s12249-022-02421-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 09/09/2022] [Indexed: 11/30/2022] Open
Abstract
Over the past few decades, co-amorphous solids have been used as a promising approach for delivering poorly water-soluble drugs. Co-amorphous solids, comprising pharmacologically relevant drug substances or excipients, improve physical stability, solubility, dissolution, and bioavailability compared with single amorphous ingredients. In this review, we have summarized recent advances in physical stability and in vitro and in vivo performances of co-amorphous solids. We have highlighted the role of molar ratio, molecular interaction, and mobility that affects the physical stability of co-amorphous solids. This review delves deep as to how co-amorphous solids affect the physicochemical properties in vitro and in vivo. We also described the challenges to the formulation of co-amorphous solids. A better understanding of the mechanisms of the physical stability, in vitro and in vivo performance of co-amorphous solids, and proper selection of the co-former is likely to expedite the development of robust co-amorphous-based pharmaceutical formulations and can address the challenges associated with the delivery of poorly soluble drugs.
Collapse
Affiliation(s)
- Qin Shi
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng, 224005, China.
| | - Yanan Wang
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng, 224005, China
| | - Sakib M Moinuddin
- College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, California, 95757, USA.,East Bay Institute For Research & Education (EBIRE), 10535 Hospital Way, Mather, California, 95655, USA
| | - Xiaodong Feng
- College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, California, 95757, USA
| | - Fakhrul Ahsan
- College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, California, 95757, USA. .,East Bay Institute For Research & Education (EBIRE), 10535 Hospital Way, Mather, California, 95655, USA.
| |
Collapse
|
7
|
Kilpeläinen T, Ervasti T, Uurasjärvi E, Koistinen A, Ketolainen J, Korhonen O, Pajula K. Detecting different amorphous - amorphous phase separation patterns in co-amorphous mixtures with high resolution imaging FTIR spectroscopy. Eur J Pharm Biopharm 2022; 180:161-169. [PMID: 36122786 DOI: 10.1016/j.ejpb.2022.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/02/2022] [Accepted: 09/12/2022] [Indexed: 11/04/2022]
Abstract
Many active pharmaceutical ingredients (API) in development suffer from low aqueous solubilities. Instead of the crystal form, the amorphous state can be used to improve the API's apparent solubility. However, the amorphous state has a higher Gibb's free energy and is inherently unstable and tends to transform back to the more stable crystal form. In co-amorphous mixtures, phase separation needs to occur before there can be crystallization. The aim of this study was to devise a method to study amorphous-amorphous phase separation with high resolution imaging Fourier transform infrared (FTIR) spectroscopy with seven 1:1 molar ratio API-API binary mixtures being examined. The binary mixtures were amorphized by melt-quenching and stored above their glass transition temperature (Tg) to monitor their phase separation. Thermodynamic properties (crystallization tendency, melting point (Tm) and Tg) of these mixtures were measured with differential scanning calorimetry (DSC) to verify the amorphization method and to assess the optimal storage temperature. The phase separation was examined with FTIR imaging in the transmission mode. Furthermore, measurements with two pure APIs were performed to ensure that the alterations occurring in the spectra were caused by phase separation not storage stress. In addition, the reproducibility of the imaging FTIR spectrometer was verified. The spectra were analyzed with principal component analysis (PCA) and a characteristic peak comparison method. Scatter-plots were produced from the amount of phase separated pixels in the measurement area as a way of visualizing the progress of phase separation. The results indicated that imaging with FTIR spectroscopy can produce reproducible results and the progress of phase separation can be detected as either a sigmoidal or as a start-to-finish linear pattern depending on the substances.
Collapse
Affiliation(s)
- Tuomas Kilpeläinen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| | - Tuomas Ervasti
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Emilia Uurasjärvi
- SIB Labs, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Arto Koistinen
- SIB Labs, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Jarkko Ketolainen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Ossi Korhonen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Katja Pajula
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| |
Collapse
|
8
|
Abramov YA, Sun G, Zeng Q. Emerging Landscape of Computational Modeling in Pharmaceutical Development. J Chem Inf Model 2022; 62:1160-1171. [PMID: 35226809 DOI: 10.1021/acs.jcim.1c01580] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Computational chemistry applications have become an integral part of the drug discovery workflow over the past 35 years. However, computational modeling in support of drug development has remained a relatively uncharted territory for a significant part of both academic and industrial communities. This review considers the computational modeling workflows for three key components of drug preclinical and clinical development, namely, process chemistry, analytical research and development, as well as drug product and formulation development. An overview of the computational support for each step of the respective workflows is presented. Additionally, in context of solid form design, special consideration is given to modern physics-based virtual screening methods. This covers rational approaches to polymorph, coformer, counterion, and solvent virtual screening in support of solid form selection and design.
Collapse
Affiliation(s)
- Yuriy A Abramov
- XtalPi, Inc., 245 Main St., Cambridge, Massachusetts 02142, United States.,Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Guangxu Sun
- XtalPi, Inc., Shenzhen Jingtai Technology Co., Ltd., Floor 3, Sf Industrial Plant, No. 2 Hongliu road, Fubao Community, Fubao Street, Futian District, Shenzhen 518100, China
| | - Qun Zeng
- XtalPi, Inc., Shenzhen Jingtai Technology Co., Ltd., Floor 3, Sf Industrial Plant, No. 2 Hongliu road, Fubao Community, Fubao Street, Futian District, Shenzhen 518100, China
| |
Collapse
|
9
|
R Sá M, Sarraguça JMG, de Sousa FF, Sarraguça MSC, Lopes JA, Lima ADDSG, Lage MR, Ribeiro PRS. Structural, thermal, vibrational, solubility and DFT studies of a tolbutamide co-amorphous drug delivery system for treatment of diabetes. Int J Pharm 2022; 615:121500. [PMID: 35077862 DOI: 10.1016/j.ijpharm.2022.121500] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 01/12/2022] [Accepted: 01/18/2022] [Indexed: 11/16/2022]
Abstract
Among the strategies for bioavailability improvement of poorly soluble drugs, co-amorphous systems have revealed to have a significant impact in the increase of the aqueous solubility of the drug, and at the same time increasing the amorphous state stability and dissolution rate when compared with the neat drug. Tolbutamide (TBM) is an oral hypoglycemic drug largely used in the treatment of type II Mellitus diabetes. TBM is a class II drug according to the Biopharmaceutical Classification System, meaning that it has low solubility and higher permeability. The aim of this study was to synthesize a co-amorphous material of tolbutamide (TBM) with tromethamine (TRIS). Density functional theory (DFT), allowed to study the structural, electronic, and thermodynamic properties, as well as solvation effects. In same theory level, several interactions tests were performed to obtain the most thermodynamically favorable drug-coformer intermolecular interactions. The vibrational spectra (mid infrared and Raman spectroscopy) are in accordance with the theoretical studies, showing that the main molecular interactions are due to the carbonyl, sulfonyl, and amide groups of TMB and the alcohol and amine groups of TRIS. X-ray powder diffraction was used to study the physical stability in dry condition at 25 °C of the co-amorphous system, indicating that the material remained in an amorphous state up to 90 days. Differential scanning calorimetry and thermogravimetric results showed a high increase of the Tg when compared with the amorphous neat drug, from 4.3 °C to 83.7 °C, which generally translated into good physical stability. Solubilities studies demonstrated an increase in the solubility of TBM by 2.5 fold when compared with its crystalline counterpart.
Collapse
Affiliation(s)
- Mônica R Sá
- Programa de Pós-Graduação em Ciências dos Materiais, Centro de Ciências Sociais, Saúde e Tecnologia, Universidade Federal do Maranhão, Imperatriz, MA 65900-410, Brazil
| | - Jorge M G Sarraguça
- Programa de Pós-Graduação em Ciências dos Materiais, Centro de Ciências Sociais, Saúde e Tecnologia, Universidade Federal do Maranhão, Imperatriz, MA 65900-410, Brazil
| | - Francisco F de Sousa
- Programa de Pós-Graduação em Ciências dos Materiais, Centro de Ciências Sociais, Saúde e Tecnologia, Universidade Federal do Maranhão, Imperatriz, MA 65900-410, Brazil; Instituto de Ciências Exatas e Naturais, Universidade Federal do Pará, CEP 66075-110 Belém, PA, Brazil
| | - Mafalda S C Sarraguça
- LAQV/REQUIMTE, Laboratório de Química Aplicada, Faculdade de Farmácia, Universidade do Porto, Portugal
| | - João A Lopes
- iMed.ULisboa, Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Antonio Douglas da S G Lima
- Programa de Pós-Graduação em Ciências dos Materiais, Centro de Ciências Sociais, Saúde e Tecnologia, Universidade Federal do Maranhão, Imperatriz, MA 65900-410, Brazil
| | - Mateus R Lage
- Programa de Pós-Graduação em Ciências dos Materiais, Centro de Ciências Sociais, Saúde e Tecnologia, Universidade Federal do Maranhão, Imperatriz, MA 65900-410, Brazil; Coordenação do Curso de Ciência e Tecnologia, Universidade Federal do Maranhão, 65800-000 Balsas, MA, Brazil
| | - Paulo R S Ribeiro
- Programa de Pós-Graduação em Ciências dos Materiais, Centro de Ciências Sociais, Saúde e Tecnologia, Universidade Federal do Maranhão, Imperatriz, MA 65900-410, Brazil.
| |
Collapse
|
10
|
Zhao X, Cheng S, Koh YP, Kelly BD, McKenna GB, Simon SL. Prediction of the Synergistic Glass Transition Temperature of Coamorphous Molecular Glasses Using Activity Coefficient Models. Mol Pharm 2021; 18:3439-3451. [PMID: 34313449 DOI: 10.1021/acs.molpharmaceut.1c00353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The glass transition temperature (Tg) of a binary miscible mixture of molecular glasses, termed a coamorphous glass, is often synergistically increased over that expected for an athermal mixture due to the strong interactions between the two components. This synergistic interaction is particularly important for the formulation of coamorphous pharmaceuticals since the molecular interactions and resulting Tg strongly impact stability against crystallization, dissolution kinetics, and bioavailability. Current models that describe the composition dependence of Tg for binary systems, including the Gordon-Taylor, Fox, Kwei, and Braun-Kovacs equations, fail to describe the behavior of coamorphous pharmaceuticals using parameters consistent with experimental ΔCP and Δα. Here, we develop a robust thermodynamic approach extending the Couchman and Karasz method through the use of activity coefficient models, including the two-parameter Margules, non-random-two-liquid (NRTL), and three-suffix Redlich-Kister models. We find that the models, using experimental values of ΔCP and fitting parameters related to the binary interactions, successfully describe observed synergistic elevations and inflections in the Tg versus composition response of coamorphous pharmaceuticals. Moreover, the predictions from the NRTL model are improved when the association-NRTL version of that model is used. Results are reported and discussed for four different coamorphous systems: indomethacin-glibenclamide, indomethacin-arginine, acetaminophen-indomethacin, and fenretinide-cholic acid.
Collapse
Affiliation(s)
- Xiao Zhao
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas 79409, United States
| | - Sixue Cheng
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas 79409, United States
| | - Yung P Koh
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas 79409, United States
| | - Brandon D Kelly
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas 79409, United States
| | - Gregory B McKenna
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas 79409, United States.,Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Sindee L Simon
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas 79409, United States.,Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| |
Collapse
|
11
|
Valkama E, Haluska O, Lehto VP, Korhonen O, Pajula K. Production and stability of amorphous solid dispersions produced by a Freeze-drying method from DMSO. Int J Pharm 2021; 606:120902. [PMID: 34293468 DOI: 10.1016/j.ijpharm.2021.120902] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 01/17/2023]
Abstract
Freeze drying is known to be able to produce an amorphous product, but this approach has been mostly used with water-based media. With APIs which are virtually water insoluble, a more appropriate freeze-drying medium would be an organic solvent. Little is known about this approach in terms of forming a stable freeze-dried amorphous product stabilized by small molecule excipient out of organic solvents. In the present study, freeze-drying of APIs from DMSO solutions was used to produce stable solid dispersions from binary mixtures of APIs containing at least one poorly water soluble or practically water-insoluble API. The developed freeze-drying method produced amorphous binary solid dispersions which remained amorphous for at least two days while the 13 best binary dispersions remained stable at room temperature for the entire study period of 127 days. Average residual DMSO levels in dried dispersions were 3.5% ± 1.6%. The developed method proved feasible in producing relatively stable amorphous solid dispersions from practically water insoluble drug compounds which could subsequently be used in further research purposes.
Collapse
Affiliation(s)
- Eetu Valkama
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Ondřej Haluska
- Department of Applied Physics, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Vesa-Pekka Lehto
- Department of Applied Physics, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Ossi Korhonen
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Katja Pajula
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
12
|
Yarlagadda DL, Sai Krishna Anand V, Nair AR, Navya Sree KS, Dengale SJ, Bhat K. Considerations for the selection of co-formers in the preparation of co-amorphous formulations. Int J Pharm 2021; 602:120649. [PMID: 33915186 DOI: 10.1016/j.ijpharm.2021.120649] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 10/21/2022]
Abstract
Co-amorphous drug delivery systems are evolving as a credible alternative to amorphous solid dispersions technology. In Co-amorphous systems (CAMs), a drug is stabilized in amorphous form using small molecular weight compounds called as co-formers. A wide variety of small molecular weight co-formers have been leveraged in the preparation of CAMs. The stability and supersaturation potential of prepared co-amorphous phases largely depend on the type of co-former employed in the CAMs. However, the rationality behind the co-former selection in co-amorphous systems is poorly understood and scarcely compiled in the literature. There are various facets to the rational selection of co-former for CAMs. In this context, the present review compiles various factors affecting the co-former selection. The factors have been broadly classified under Thermodynamic, Kinetic and Pharmacokinetic-Pharmacologically relevant parameters. In particular, the importance of Glass transition, Miscibility, Liquid-Liquid phase separation (LLPS), Crystallization inhibition has been deliberated in detail.
Collapse
Affiliation(s)
- Dani Lakshman Yarlagadda
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576 104, India
| | - Vullendula Sai Krishna Anand
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576 104, India
| | - Athira R Nair
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576 104, India
| | - K S Navya Sree
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576 104, India
| | - Swapnil J Dengale
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576 104, India
| | - Krishnamurthy Bhat
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576 104, India.
| |
Collapse
|
13
|
Co-Amorphous Drug Formulations in Numbers: Recent Advances in Co-Amorphous Drug Formulations with Focus on Co-Formability, Molar Ratio, Preparation Methods, Physical Stability, In Vitro and In Vivo Performance, and New Formulation Strategies. Pharmaceutics 2021; 13:pharmaceutics13030389. [PMID: 33804159 PMCID: PMC7999207 DOI: 10.3390/pharmaceutics13030389] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/20/2022] Open
Abstract
Co-amorphous drug delivery systems (CAMS) are characterized by the combination of two or more (initially crystalline) low molecular weight components that form a homogeneous single-phase amorphous system. Over the past decades, CAMS have been widely investigated as a promising approach to address the challenge of low water solubility of many active pharmaceutical ingredients. Most of the studies on CAMS were performed on a case-by-case basis, and only a few systematic studies are available. A quantitative analysis of the literature on CAMS under certain aspects highlights not only which aspects have been of great interest, but also which future developments are necessary to expand this research field. This review provides a comprehensive updated overview on the current published work on CAMS using a quantitative approach, focusing on three critical quality attributes of CAMS, i.e., co-formability, physical stability, and dissolution performance. Specifically, co-formability, molar ratio of drug and co-former, preparation methods, physical stability, and in vitro and in vivo performance were covered. For each aspect, a quantitative assessment on the current status was performed, allowing both recent advances and remaining research gaps to be identified. Furthermore, novel research aspects such as the design of ternary CAMS are discussed.
Collapse
|
14
|
Abstract
Co-amorphous (CAM) systems are promising drug-delivery systems in the arena of therapeutic drug delivery, addressing the poor aqueous solubility of drugs by enhancing solubility and thereby improving the oral bioavailability and therapeutic effect of the drug. A CAM system is a single-phase homogeneous blend of two or more low molecular weight molecules that can be drug–drug or drug–co-former, stabilized via intermolecular interactions, adding the benefit of thermodynamic stability. This review covers the fundamentals of CAM systems and recent advances in formulation development. In particular, we strive to address the theoretical, molecular, technical and biopharmaceutical aspects, advantages over polymeric amorphous solid dispersions, mechanisms of stabilization of amorphous forms, insights into unexplored in silico tools in excipient selection and regulatory viewpoints.
Collapse
|
15
|
Yu D, Kan Z, Shan F, Zang J, Zhou J. Triple Strategies to Improve Oral Bioavailability by Fabricating Coamorphous Forms of Ursolic Acid with Piperine: Enhancing Water-Solubility, Permeability, and Inhibiting Cytochrome P450 Isozymes. Mol Pharm 2020; 17:4443-4462. [PMID: 32926628 DOI: 10.1021/acs.molpharmaceut.0c00443] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
As a BCS IV drug, ursolic acid (UA) has low oral bioavailability mainly because of its poor aqueous solubility/dissolution, poor permeability, and metabolism by cytochrome P450 (CYP) isozymes, such as CYP3A4. Most UA preparations demonstrated a much higher dissolution than that of its crystalline form yet a low drug concentration in plasma due to their lower consideration or evaluation for the permeability and metabolism issues. In the current study, a supramolecular coamorphous system of UA with piperine (PIP) was prepared and characterized by powder X-ray diffraction, differential scanning calorimetry, and scanning electron microscopy. In comparison to crystalline UA and UA in physical mixture, such coamorphous system enhanced solubility (5.3-7-fold in the physiological solution) and dissolution (7-8-fold in the physiological solution within 2 h) of UA and exhibited excellent physical stability under 90-day storage conditions. More importantly, the pharmacokinetic study of coamorphous UA in rats exhibited 5.8-fold and 2.47-fold improvement in AUC0-∞ value, respectively, compared with its free and mixed crystalline counterparts. In order to further explore the mechanism of such improvement, the molecular interactions of a coamorphous system in the solid state were investigated. Fourier transform infrared spectroscopy, solid-state 13C nuclear magnetic resonance spectroscopy, and density functional theory modeling suggested that intermolecular hydrogen bonds with strong interactions newly formed between UA and PIP after coamorphization. The in vitro permeability studies across Caco-2 cell monolayer and metabolism studies by rat hepatic microsomes indicated that free PIP significantly increased the permeability of UA and inhibited the enzymatic metabolism of UA by CYP3A4. However, PIP in the coamorphous combination exhibited a much lower level in the bioenhancing than its free form arising from the synchronized dissolution characteristic of the preparation (only 60% of PIP released in comparison to its free counterpart in 2 h). The in situ loop study in rats proposed that the acid-sensitive dissolution in the stomach of the coamorphous preparation helped to improve the effective free drug concentration, thereby facilitating PIP to play its role in bioenhancing. The current study offers an exploratory strategy to overcome poor solubility/dissolution, poor permeability, and metabolism by cytochrome P450 isozymes of the BCS IV drug to improve its oral bioavailability.
Collapse
Affiliation(s)
- Danni Yu
- Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing 211198, PR China
| | - Zigui Kan
- Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing 211198, PR China
- School of Chemistry and Chemical Engineering, Key Laboratory of Mesoscopic Chemistry of MOE, Nanjing University, Nanjing 210093, PR China
| | - Fei Shan
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Jing Zang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Jianping Zhou
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| |
Collapse
|
16
|
Han J, Wei Y, Lu Y, Wang R, Zhang J, Gao Y, Qian S. Co-amorphous systems for the delivery of poorly water-soluble drugs: recent advances and an update. Expert Opin Drug Deliv 2020; 17:1411-1435. [DOI: 10.1080/17425247.2020.1796631] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Jiawei Han
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Yuanfeng Wei
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Yan Lu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Runze Wang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Jianjun Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Yuan Gao
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Shuai Qian
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| |
Collapse
|
17
|
Raman imaging of amorphous-amorphous phase separation in small molecule co-amorphous systems. Eur J Pharm Biopharm 2020; 155:49-54. [PMID: 32795500 DOI: 10.1016/j.ejpb.2020.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/01/2020] [Accepted: 08/09/2020] [Indexed: 11/21/2022]
Abstract
Many new active pharmaceutical ingredients (API) undergoing development have low permeabilities or low aqueous solubilities. However, the amorphous state is usually more soluble than its crystalline counterpart. The amorphous state has a higher Gibb's free energy, which can improve the apparent solubility but decrease the stability since the amorphous state tends to transform to the more stable crystalline form. Before recrystallization, a co-amorphous binary mixture's ingredients have to undergo a phase separation. The aim of this study was to obtain a better understanding of the amorphous-amorphous phase separation in co-amorphous binary mixtures and test the suitability of imaging Raman spectroscopy for detecting this phenomenon. To study the phase separation, we prepared three different 50:50 mass ratio binary mixtures of APIs: paracetamol-terfenadine, (PAR-TRF), paracetamol-indomethacin (PAR-IMC) and terfenadine-indomethacin (TRF-IMC). The binary mixtures were amorphized with melt-quenching and stored above their glass transition temperature (Tg) to monitor their phase separation. Thermal degradation was determined with a high performance liquid chromatography (HPLC) method to ensure that melt-quenching did not cause any thermal degradation of the molecules. Thermodynamic attributes (crystallization tendency, melting point (Tm) and Tg) were measured with differential scanning calorimetry (DSC) to ensure that the co-amorphous systems transformed to the amorphous state and remained amorphous after cooling and reheating. Phase separation was studied from the surface and cross-section (CS) with Raman imaging to examine if it occurred more on the surface than in the bulk. The Raman spectra were analyzed with principal component analysis (PCA) and Contour plots were produced from the PCA-score values to visualize concentration differences in the mixtures. The results showed that API vs API concentrations increased as a function of time in both surface and CS images before crystallization. This suggests that Raman imaging is a suitable technique to detect the phase separation phenomena in small molecule co-amorphous binary mixtures.
Collapse
|
18
|
Pajula K, Hyyryläinen J, Koistinen A, Leskinen JT, Korhonen O. Detection of amorphous-amorphous phase separation in small molecular co-amorphous mixtures with SEM-EDS. Eur J Pharm Biopharm 2020; 150:43-49. [DOI: 10.1016/j.ejpb.2020.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/28/2020] [Accepted: 03/03/2020] [Indexed: 10/24/2022]
|
19
|
Molecular Dynamics and Physical Stability of Pharmaceutical Co-amorphous Systems: Correlation Between Structural Relaxation Times Measured by Kohlrausch-Williams-Watts With the Width of the Glass Transition Temperature (ΔT g) and the Onset of Crystallization. J Pharm Sci 2019; 108:3848-3858. [PMID: 31542436 DOI: 10.1016/j.xphs.2019.09.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 08/18/2019] [Accepted: 09/11/2019] [Indexed: 11/20/2022]
Abstract
The study aims to characterize the structural relaxation times of quench-cooled co-amorphous systems using Kohlrausch-Williams-Watts (KWW) and to correlate the relaxation data with the onset of crystallization. Comparison was also made between the relaxation times obtained by KWW and the width of glass transition temperature (ΔTg) methods (simple and quick). Differential scanning calorimetry, Fourier-transformed infrared spectroscopy, and polarized light microscopy were used to characterize the systems. Results showed that co-amorphous systems yielded a single Tg and ΔCp, suggesting the binary mixtures exist as a single amorphous phase. A narrow step change at Tg indicates the systems were fragile glasses. In co-amorphous nap-indo and para-indo, experimental Tgs were in good agreement with the predicted Tg. However, the Tg of co-amorphous nap-cim and indo-cim were 20°C higher than the predicted Tg, possibly due to stronger molecular interactions. Structural relaxation times below the experimental Tg were successfully characterized using the KWW and ΔTg methods. The comparison plot showed that KWW data are directly proportional to the ½ power of ΔTg data, after adjusting for a small offset. A moderate positive correlation was observed between the onset of crystallization and the KWW data. Structural relaxation times may be useful predictor of physical stability of co-amorphous systems.
Collapse
|
20
|
Shi Q, Moinuddin SM, Cai T. Advances in coamorphous drug delivery systems. Acta Pharm Sin B 2019; 9:19-35. [PMID: 30766775 PMCID: PMC6361732 DOI: 10.1016/j.apsb.2018.08.002] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/11/2018] [Accepted: 08/12/2018] [Indexed: 01/18/2023] Open
Abstract
In recent years, the coamorphous drug delivery system has been established as a promising formulation approach for delivering poorly water-soluble drugs. The coamorphous solid is a single-phase system containing an active pharmaceutical ingredient (API) and other low molecular weight molecules that might be pharmacologically relevant APIs or excipients. These formulations exhibit considerable advantages over neat crystalline or amorphous material, including improved physical stability, dissolution profiles, and potentially enhanced therapeutic efficacy. This review provides a comprehensive overview of coamorphous drug delivery systems from the perspectives of preparation, physicochemical characteristics, physical stability, in vitro and in vivo performance. Furthermore, the challenges and strategies in developing robust coamorphous drug products of high quality and performance are briefly discussed.
Collapse
Key Words
- API, active pharmaceutical ingredient;
- AUC, area under plasma concentrations-time curve
- BCS, bio-pharmaceutics classification systems
- Bioavailability
- Characterization
- Cmax, maximum plasma concentration
- Coamorphous
- Css, plasma concentration at steady state
- DSC, differential scanning calorimetry
- DVS, dynamic vapor sorption
- Dc, relative degree of crystallization
- Dissolution
- FT-IR, fourier transform infrared spectroscopy
- HME, hot melt extrusion
- HPLC, high performance liquid chromatography
- IDR, intrinsic dissolution rate
- LFRS, low-frequency Raman spectroscopy
- LLPS, liquid—liquid phase separation
- MTDSC, modulated temperature differential scanning calorimetry
- NMR, nuclear magnetic resonance
- P-gp, P-glycoprotein
- PXRD, powder X-ray diffraction
- Physical stability
- Preparation
- RH, relative humidity
- SEM, scanning electron microscope
- TGA, thermogravimetric analysis
- Tg, glass transition temperature
- Tmax, time of maximum plasma concentration
- UV, ultraviolet spectroscopy
Collapse
Affiliation(s)
| | | | - Ting Cai
- Corresponding author. Tel.: +86 25 83271123.
| |
Collapse
|
21
|
Karagianni A, Kachrimanis K, Nikolakakis I. Co-Amorphous Solid Dispersions for Solubility and Absorption Improvement of Drugs: Composition, Preparation, Characterization and Formulations for Oral Delivery. Pharmaceutics 2018; 10:pharmaceutics10030098. [PMID: 30029516 PMCID: PMC6161132 DOI: 10.3390/pharmaceutics10030098] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 02/06/2023] Open
Abstract
The amorphous solid state offers an improved apparent solubility and dissolution rate. However, due to thermodynamic instability and recrystallization tendencies during processing, storage and dissolution, their potential application is limited. For this reason, the production of amorphous drugs with adequate stability remains a major challenge and formulation strategies based on solid molecular dispersions are being exploited. Co-amorphous systems are a new formulation approach where the amorphous drug is stabilized through strong intermolecular interactions by a low molecular co-former. This review covers several topics applicable to co-amorphous drug delivery systems. In particular, it describes recent advances in the co-amorphous composition, preparation and solid-state characterization, as well as improvements of dissolution performance and absorption are detailed. Examples of drug-drug, drug-carboxylic acid and drug-amino acid co-amorphous dispersions interacting via hydrogen bonding, π−π interactions and ionic forces, are presented together with corresponding final dosage forms.
Collapse
Affiliation(s)
- Anna Karagianni
- Department of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| | - Kyriakos Kachrimanis
- Department of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| | - Ioannis Nikolakakis
- Department of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| |
Collapse
|
22
|
Meng-Lund H, Kasten G, Jensen KT, Poso A, Pantsar T, Rades T, Rantanen J, Grohganz H. The use of molecular descriptors in the development of co-amorphous formulations. Eur J Pharm Sci 2018; 119:31-38. [DOI: 10.1016/j.ejps.2018.04.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/21/2018] [Accepted: 04/08/2018] [Indexed: 11/17/2022]
|
23
|
Tian Y, Jones DS, Donnelly C, Brannigan T, Li S, Andrews GP. A New Method of Constructing a Drug–Polymer Temperature–Composition Phase Diagram Using Hot-Melt Extrusion. Mol Pharm 2017; 15:1379-1391. [DOI: 10.1021/acs.molpharmaceut.7b00445] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Yiwei Tian
- Pharmaceutical Engineering Group, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - David S. Jones
- Pharmaceutical Engineering Group, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Conor Donnelly
- Pharmaceutical Engineering Group, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Timothy Brannigan
- Pharmaceutical Engineering Group, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Shu Li
- Pharmaceutical Engineering Group, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Gavin P. Andrews
- Pharmaceutical Engineering Group, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| |
Collapse
|
24
|
Newman A, Reutzel-Edens SM, Zografi G. Coamorphous Active Pharmaceutical Ingredient-Small Molecule Mixtures: Considerations in the Choice of Coformers for Enhancing Dissolution and Oral Bioavailability. J Pharm Sci 2017; 107:5-17. [PMID: 28989014 DOI: 10.1016/j.xphs.2017.09.024] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 09/19/2017] [Accepted: 09/22/2017] [Indexed: 10/18/2022]
Abstract
In the recent years, coamorphous systems, containing an active pharmaceutical ingredient (API) and a small molecule coformer have appeared as alternatives to the use of either amorphous solid dispersions containing polymer or cocrystals of API and small molecule coformers, to improve the dissolution and oral bioavailability of poorly soluble crystalline API. This Commentary article considers the relative properties of amorphous solid dispersions and coamorphous systems in terms of methods of preparation; miscibility; glass transition temperature; physical stability; hygroscopicity; and aqueous dissolution. It also considers important questions concerning the fundamental criteria to be used for the proper selection of a small molecule coformer regarding its ability to form either coamorphous or cocrystal systems. Finally, we consider various aspects of product development that are specifically associated with the formulation of commercial coamorphous systems as solid oral dosage forms. These include coformer selection; screening; methods of preparation; preformulation; physical stability; bioavailability; and final formulation. Through such an analysis of coamorphous API-small molecule coformer systems, against the more widely studied API-polymer dispersions and cocrystals, it is believed that the strengths and weaknesses of coamorphous systems can be better understood, leading to more efficient formulation and manufacture of such systems for enhancing oral bioavailability.
Collapse
Affiliation(s)
- Ann Newman
- Seventh Street Development Group LLC, Kure Beach, North Carolina 28449.
| | - Susan M Reutzel-Edens
- Small Molecule Design and Development, Eli Lilly and Company, Indianapolis, Indiana 46285
| | - George Zografi
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53706
| |
Collapse
|
25
|
Moriyama K, Yasuhara Y, Ota H. Visualization of Protonation/Deprotonation of Active Pharmaceutical Ingredient in Solid State by Vapor Phase Amine-Selective Alkyne Tagging and Raman Imaging. J Pharm Sci 2017; 106:1778-1785. [DOI: 10.1016/j.xphs.2017.03.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 02/28/2017] [Accepted: 03/06/2017] [Indexed: 01/05/2023]
|
26
|
Correlation between calculated molecular descriptors of excipient amino acids and experimentally observed thermal stability of lysozyme. Int J Pharm 2017; 523:238-245. [DOI: 10.1016/j.ijpharm.2017.03.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 02/08/2017] [Accepted: 03/19/2017] [Indexed: 11/21/2022]
|
27
|
DeBoyace K, Wildfong PLD. The Application of Modeling and Prediction to the Formation and Stability of Amorphous Solid Dispersions. J Pharm Sci 2017; 107:57-74. [PMID: 28389266 DOI: 10.1016/j.xphs.2017.03.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 03/27/2017] [Indexed: 02/06/2023]
Abstract
Amorphous solid dispersion (ASD) formulation development is frequently difficult owing to the inherent physical instability of the amorphous form, and limited understanding of the physical and chemical interactions that translate to initial dispersion formation and long-term physical stability. Formulation development for ASDs has been historically accomplished through trial and error or experience with extant systems; however, rational selection of appropriate excipients is preferred to reduce time to market and decrease costs associated with development. Current efforts to develop thermodynamic and computational models attempt to rationally direct formulation and show promise. This review compiles and evaluates important methods used to predict ASD formation and physical stability. Recent literature in which these methods are applied is also reviewed, and limitations of each method are discussed.
Collapse
Affiliation(s)
- Kevin DeBoyace
- Department of Pharmaceutical Sciences, Duquesne University, 600 Forbes Av, Pittsburgh, Pennsylvania 15282
| | - Peter L D Wildfong
- Department of Pharmaceutical Sciences, Duquesne University, 600 Forbes Av, Pittsburgh, Pennsylvania 15282.
| |
Collapse
|
28
|
Co amorphous systems: A product development perspective. Int J Pharm 2016; 515:403-415. [DOI: 10.1016/j.ijpharm.2016.10.043] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 10/18/2016] [Accepted: 10/19/2016] [Indexed: 11/21/2022]
|
29
|
Korhonen O, Pajula K, Laitinen R. Rational excipient selection for co-amorphous formulations. Expert Opin Drug Deliv 2016; 14:551-569. [DOI: 10.1080/17425247.2016.1198770] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Ossi Korhonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Katja Pajula
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Riikka Laitinen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
30
|
Dengale SJ, Grohganz H, Rades T, Löbmann K. Recent advances in co-amorphous drug formulations. Adv Drug Deliv Rev 2016; 100:116-25. [PMID: 26805787 DOI: 10.1016/j.addr.2015.12.009] [Citation(s) in RCA: 300] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 12/09/2015] [Indexed: 11/19/2022]
Abstract
Co-amorphous drug delivery systems have recently gained considerable interest in the pharmaceutical field because of their potential to improve oral bioavailability of poorly water-soluble drugs through drug dissolution enhancement as a result of the amorphous nature of the material. A co-amorphous system is characterized by the use of only low molecular weight components that are mixed into a homogeneous single-phase co-amorphous blend. The use of only low molecular weight co-formers makes this approach very attractive, as the amount of amorphous stabilizer can be significantly reduced compared with other amorphous stabilization techniques. Because of this, several research groups started to investigate the co-amorphous formulation approach, resulting in an increasing amount of scientific publications over the last few years. This study provides an overview of the co-amorphous field and its recent findings. In particular, we investigate co-amorphous formulations from the viewpoint of solid dispersions, describe their formation and mechanism of stabilization, study their impact on dissolution and in vivo performance and briefly outline the future potentials.
Collapse
Affiliation(s)
- Swapnil Jayant Dengale
- Manipal College of Pharmaceutical Sciences, Manipal University, Manipal, Karnataka, India
| | - Holger Grohganz
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Rades
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark.
| | - Korbinian Löbmann
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
31
|
Huang Y, Zhang Q, Wang JR, Lin KL, Mei X. Amino acids as co-amorphous excipients for tackling the poor aqueous solubility of valsartan. Pharm Dev Technol 2016; 22:69-76. [DOI: 10.3109/10837450.2016.1163390] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Ying Huang
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Qi Zhang
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jian-Rong Wang
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Kai-Lei Lin
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xuefeng Mei
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|