1
|
Shetty N, Hau J, Tang S, Chiang PC, Liu J, Jia W, Lubach JW, Nagapudi K, Hou HH. Assessing the impacts of drug loading and polymer type on dissolution behavior and diffusive flux of GDC-6893 amorphous solid dispersions. J Pharm Sci 2025; 114:103686. [PMID: 39880163 DOI: 10.1016/j.xphs.2025.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/12/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025]
Abstract
It is desirable but remains challenging to develop high drug load amorphous solid dispersions (ASDs) without compromising their quality attributes and bio-performance. In this work, we investigated the impacts of formulation variables, such as drug loading (DL) and polymer type, on dissolution behavior, diffusive flux, and in vitro drug absorption of ASDs of a high Tg compound, GDC-6893. ASDs with two polymers (HPMCAS and PVPVA) and various DLs (20 - 80%) were produced by spray drying and their drug-polymer miscibility was evaluated using solid-state nuclear magnetic resonance (ssNMR). μFLUX™ apparatus was used to evaluate the dissolution and drug membrane transport of ASDs at target solution concentrations above the amorphous solubility. Polymer release was monitored using a high-performance liquid chromatography (HPLC) equipped with a charged aerosol detector (CAD). Subsequently, bio-accessibility (%BioA) profiles of the ASDs were evaluated using a benchtop Gastro-Intestinal Model with an advanced gastric compartment (Tiny-TIM), capable of simulating the GI transit as well as in vitro drug dissolution and absorption. Good miscibility and physical stability were observed in ASDs with both HPMCAS and PVPVA even at a high DL of 80%. All GDC-6893 ASDs exhibited dissolution profiles surpassing the amorphous solubility of 20 µg/mL, regardless of the DL and the type of polymer used. Glass-liquid phase separation (GLPS) was observed for ASDs, even at the DL of 80%, and all of these systems reached the maximum achievable diffusive flux. Tiny-TIM results showed an improvement in the %BioA of GDC-6893 ASD compared to its crystalline counterpart however the drug loading and polymer type had no significant impacts on %BioA profiles. Insights from this study suggest that although congruent drug and polymer release was not observed for both HMPCAS- and PVPVA-based ASDs at both 20% and 80% DLs, GDC-6893 and the polymer (HPMCAS or PVPVA) dissolved rapidly from high DL ASDs, followed by the occurrence of GLPS, resulting in the formation of nanosized colloidal species. The findings described herein highlight the importance of understanding both drug and polymer dissolution behavior, as well as in vitro drug absorption, which are essential for the rational design of optimal formulations with desired quality and bio-performance.
Collapse
Affiliation(s)
- Nivedita Shetty
- Department of Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jonathan Hau
- Department of Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Shijia Tang
- Department of Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Po-Chang Chiang
- Department of Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jia Liu
- Department of Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Wei Jia
- Department of Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Joseph W Lubach
- Department of Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Karthik Nagapudi
- Department of Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hao Helen Hou
- Department of Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
2
|
Kapourani A, Pantazos I, Valkanioti V, Chatzitheodoridou M, Kalogeri C, Barmpalexis P. Unveiling the impact of preparation methods, matrix/carrier type selection and drug loading on the supersaturation performance of amorphous solid dispersions. Int J Pharm 2025; 671:125242. [PMID: 39842744 DOI: 10.1016/j.ijpharm.2025.125242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/24/2025]
Abstract
Amorphous solid dispersions (ASDs) are widely recognized for their potential to enhance the solubility of poorly water-soluble drugs, with factors such as molecular mobility, intermolecular interactions, and storage conditions playing critical roles in their performance. However, the influence of preparation methods on their performance remains underexplored, especially regarding their supersaturation . To address this gap, the present study systematically investigates ASDs of ibuprofen (IBU, used as a model drug) prepared using two widely utilized techniques (solvent evaporation, SE, and melt-quench cooling, M-QC). Three different matrices/carriers (Soluplus®, SOL, povidone, PVP, and copovidone, PVPVA) were employed to evaluate the combined influence of preparation method, matrix/carrier type, and drug loading on ASD performance. Supersaturation behavior during dissolution, particularly its dependence on the Sink Index (SI), was a key focus. All ASDs showed successful amorphization, but molecular near-order structures differed based on the preparation method. ATR-FTIR spectroscopy revealed stronger molecular interactions in M-QC ASDs (compared to SE). Dissolution studies under supersaturation conditions (SI = 0.1 and SI = 0.2) highlighted significant performance differences. M-QC ASDs consistently exhibited higher in vitro AUC(0→t) values under non-sink conditions compared to crystalline IBU. Conversely, SE ASDs showed improved supersaturation primarily under low SI conditions, especially with SOL at low drug loadings. The findings underscore the need for a systematic approach in developing ASDs, considering preparation method, matrix/carrier type, drug loading and dissolution study conditions collectively. These factors significantly influence dissolution behavior and supersaturation, emphasizing that they should not be independently studied but evaluated comprehensively to optimize ASD performance.
Collapse
Affiliation(s)
- Afroditi Kapourani
- Laboratory of Pharmaceutical Technology, Division of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Ioannis Pantazos
- Laboratory of Pharmaceutical Technology, Division of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Vasiliki Valkanioti
- Laboratory of Pharmaceutical Technology, Division of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Melina Chatzitheodoridou
- Laboratory of Pharmaceutical Technology, Division of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Christina Kalogeri
- Laboratory of Pharmaceutical Technology, Division of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Panagiotis Barmpalexis
- Laboratory of Pharmaceutical Technology, Division of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; Natural Products Research Centre of Excellence-AUTH (NatPro-AUTH), Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Thessaloniki 57001, Greece.
| |
Collapse
|
3
|
Juneja M, Mehtre K, Saini V, Singh R, Amate P, Kashyap M, Sangamwar AT. Synergistic effect of polymers in stabilizing amorphous pretomanid through high drug loaded amorphous solid dispersion. Drug Deliv Transl Res 2025; 15:717-731. [PMID: 38837117 DOI: 10.1007/s13346-024-01630-w] [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] [Accepted: 05/13/2024] [Indexed: 06/06/2024]
Abstract
Pretomanid (PTM), an oral antibiotic used in the treatment of adults with pulmonary extensively drug-resistant, nonresponsive multidrug-resistant tuberculosis (MDR-TB). It is a poor glass former, that shows high recrystallization tendency from the amorphous and supersaturated state, resulting in low aqueous solubility and suboptimal absorption through the gastrointestinal tract. The present investigation aimed to develop high drug loaded ternary amorphous solid dispersions (ASDs) of PTM with improved stability and enhanced biopharmaceutical performance by utilizing a combination of polymers. The polymers were comprehensively screened based on drug-polymer miscibility and saturation solubility analysis. A combination of Hydroxypropyl Methylcellulose Acetate Succinate (HPMCAS-HF) and Polyvinylpyrrolidone K-30 (PVP K-30) showed synergism in drug-polymer miscibility as evidenced through pronounced depression in the melting endotherm of PTM. The Powder X-ray Diffraction (P-XRD) diffractograms of 30% w/w PTM loaded ternary ASDs displayed the halo pattern, contrary to the binary ASDs. Drug-polymer interactions (hydrophobic forces) involved between PTM and polymers were detected through Fourier Transform Infrared Spectroscopy (FT-IR) and Nuclear Magnetic Resonance Spectroscopy (13C-NMR) which contributed to the synergistic enhancement in solubility and dissolution of ternary ASDs with sustained release over 12 h. Ternary ASDs demonstrated better in-vivo performance compared to the binary ASDs, showing a 4.63-fold increase in maximum plasma concentration. All ASDs remained stable and resisted phase separation during short-term stability studies for 3 months at ambient conditions. It was concluded that the hydrophobic and hydrophilic polymeric combination (HPMCAS-HF and PVP K-30, respectively) effectively prevented the crystallization and ensured sustained drug release with improved in-vivo absorption of PTM.
Collapse
Affiliation(s)
- Mehak Juneja
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab, India, 160062
| | - Krishna Mehtre
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab, India, 160062
| | - Vanshul Saini
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab, India, 160062
| | - Ridhima Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab, India, 160062
| | - Prakash Amate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab, India, 160062
| | - Mahesh Kashyap
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab, India, 160062
| | - Abhay T Sangamwar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab, India, 160062.
| |
Collapse
|
4
|
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
|
5
|
Chakraborty S, Bansal AK. Application of atomic force microscopy in the development of amorphous solid dispersion. J Pharm Sci 2025; 114:70-81. [PMID: 39481473 DOI: 10.1016/j.xphs.2024.10.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024]
Abstract
Development of Amorphous Solid Dispersion (ASD) requires an in-depth characterization at different stages due to its structural and functional complexity. Various tools are conventionally used to investigate the processing, stability, and functionality of ASDs. However, many subtle features remain poorly understood due to lack of nano-scale characterization tools in routine practice. Atomic force microscopy (AFM) is a type of scanning probe microscopy, used for high resolution imaging and measuring features at the nano-scale. In recent years AFM has been used increasingly as a characterization tool in different areas of the development of ASD, including drug-polymer miscibility, localized characterization of the phase separated domains, lateral molecular diffusivity on ASD surface, crystallinity and crystallization kinetics in ASD, phase behavior of ASD during dissolution, and conformation of polymer during dissolution. In this review, we have highlighted the current applications of AFM in capturing critical aspects of stability and dissolution behavior of ASD. Potential areas of future development in this domain have been discussed.
Collapse
Affiliation(s)
- Soumalya Chakraborty
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-S.A.S. Nagar, Punjab 160062, India
| | - Arvind K Bansal
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-S.A.S. Nagar, Punjab 160062, India.
| |
Collapse
|
6
|
Deac A, Que C, Cousineau ML, Indulkar AS, Gao Y, Zhang GGZ, Taylor LS. Dissolution mechanisms of amorphous solid dispersions: Role of polymer molecular weight and identification of a new failure mode. J Pharm Sci 2025; 114:486-496. [PMID: 39461502 DOI: 10.1016/j.xphs.2024.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 10/29/2024]
Abstract
The mechanisms of drug release from amorphous solid dispersions (ASDs) are complex and not fully explored, making it difficult to optimize for in vivo performance. A recurring behavior has been the limit of congruency (LoC), a drug loading above which the ASD surface forms an amorphous drug-rich barrier in the presence of water, which hinders release, especially in non-sink conditions. Drug-polymer interactions and drug glass transition temperature were reported to affect the LoC. However, the effect of polymer molecular weight has not been explored. ASDs of clotrimazole and different molecular weight grades of poly (vinylpyrrolidone) (PVP) were studied for their release to obtain their LoC drug loadings. Failure modes underpinning the LoC were investigated using fluorescence confocal microscopy to analyze the ASD/solution interface and phase behavior of ASD films at high relative humidity. ASDs with good release formed stable drug-rich nanodroplets at the ASD/solution interface, while ASDs with poor release were limited by one of two failure modes, depending on PVP molecular weight. In Failure Mode I the nanodroplets quickly agglomerated, while in Failure Mode II the system underwent phase inversion. This work highlights the importance of identifying the mechanisms underlying the LoC to improve the release of higher drug loading ASDs.
Collapse
Affiliation(s)
- Alexandru Deac
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Chailu Que
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Michelle L Cousineau
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Anura S Indulkar
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, IL 60064, United States
| | - Yi Gao
- Formulation Development, Drug Product Science & Technology, AbbVie Inc., North Chicago, IL 60064, United States.
| | - Geoff G Z Zhang
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States; Development Sciences, Research and Development, AbbVie Inc., North Chicago, IL 60064, United States; ProPhysPharm LLC, Lincolnshire, IL 60069, United States
| | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
7
|
Bapat P, Schwabe R, Paul S, Tseng YC, Bergman C, Taylor LS. Exploring biorelevant conditions and release profiles of ritonavir from HPMCAS-based amorphous solid dispersions. J Pharm Sci 2025; 114:185-198. [PMID: 39186978 DOI: 10.1016/j.xphs.2024.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 08/28/2024]
Abstract
Development of a release test for amorphous solid dispersions (ASDs) that is in vivo predictive is essential to identify optimally performing formulations early in development. For ASDs containing an enteric polymer, consideration of buffer properties is essential. Herein, release rates of hydroxypropyl methyl cellulose acetate succinate (HPMCAS) and ritonavir from ASDs with a 20% drug loading were compared in phosphate and bicarbonate buffers with different molarities, at pH 6.5. The bioaccessibility of ritonavir from the ASD in the tiny-TIM apparatus was also evaluated and compared to that of the crystalline drug. The surface pH at the dissolving solid: solution interface was evaluated using a pH-sensitive fluorescence probe for HPMCAS and ASD compacts in phosphate and bicarbonate buffers. Drug and polymer were found to release congruently in all buffer systems, indicating that the polymer controlled the drug release. Release was slowest in 10 mM bicarbonate buffer, and much faster in phosphate buffers with molarities typically used in release testing (20-50 mM). Release from the 10 mM bicarbonate buffer was matched in a 5 mM phosphate buffer. The surface pH of HPMCAS and HPMCAS:ritonavir ASDs was found to be lower than the bulk solution pH, where surface pH differences largely explained release rate differences seen in the different buffer systems. Ritonavir was highly bioaccessible from the ASD, as assessed by the tiny-TIM system, and much less bioaccessible when crystalline drug was used. The observations highlight the need for continued development of biorelevant assays tailored for ASD formulation assessment.
Collapse
Affiliation(s)
- Pradnya Bapat
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Robert Schwabe
- Material and Analytical Sciences, Research and Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT 06877, United States
| | - Shubhajit Paul
- Material and Analytical Sciences, Research and Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT 06877, United States
| | - Yin-Chao Tseng
- Material and Analytical Sciences, Research and Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT 06877, United States
| | - Cameron Bergman
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
8
|
Ueda K, Moseson DE, Taylor LS. Amorphous solubility advantage: Theoretical considerations, experimental methods, and contemporary relevance. J Pharm Sci 2025; 114:18-39. [PMID: 39222748 DOI: 10.1016/j.xphs.2024.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Twenty-five years ago, Hancock and Parks asked a provocative question: "what is the true solubility advantage for amorphous pharmaceuticals?" Difficulties in determining the amorphous solubility have since been overcome due to significant advances in theoretical understanding and experimental methods. The amorphous solubility is now understood to be the concentration after the drug undergoes liquid-liquid or liquid-glass phase separation, forming a water-saturated drug-rich phase in metastable equilibrium with an aqueous phase containing molecularly dissolved drug. While crystalline solubility is an essential parameter impacting the absorption of crystalline drug formulations, amorphous solubility is a vital factor for considering absorption from supersaturating formulations. However, the amorphous solubility of drugs is complex, especially in the presence of formulation additives and gastrointestinal components, and concentration-based measurements may not indicate the maximum drug thermodynamic activity. This review discusses the concept of the amorphous solubility advantage, including a historical perspective, theoretical considerations, experimental methods for amorphous solubility measurement, and the contribution of supersaturation and amorphous solubility to drug absorption. Leveraging amorphous solubility and understanding the associated physicochemical principles can lead to more effective development strategies for poorly water-soluble drugs, ultimately benefiting therapeutic outcomes.
Collapse
Affiliation(s)
- Keisuke Ueda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Dana E Moseson
- Worldwide Research and Development, Pfizer, Inc., Groton, CT 06340, United States
| | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
9
|
Moseson DE, Li N, Rantanen J, Ueda K, Zhang GGZ. Professor Lynne S. Taylor: Scientist, educator, and adventurer. J Pharm Sci 2025; 114:2-9. [PMID: 39426563 DOI: 10.1016/j.xphs.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
This special edition of the Journal of Pharmaceutical Sciences is dedicated to Professor Lynne S. Taylor (Retter Distinguished Professor of Pharmacy, Department of Industrial and Molecular Pharmaceutics, Purdue University), to honor her distinguished career as a pharmaceutical scientist and educator. The goal of this commentary is to provide an overview of Professor Taylor's career path, summarize her key research contributions, and provide some insight into her personal and professional contributions as an educator, mentor, wife, mother, friend, and adventurer.
Collapse
Affiliation(s)
- Dana E Moseson
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States.
| | - Na Li
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Jukka Rantanen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Keisuke Ueda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Geoff G Z Zhang
- ProPhysPharm LLC, Lincolnshire, Illinois 60069, United States; Department of Industrial and Molecular Pharmaceutics, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
10
|
Horváth ZM, Petersone L, Mohylyuk V. Quantification of soluplus for dissolution tests: SEC method development and validation. J Pharm Sci 2025; 114:157-164. [PMID: 38972546 DOI: 10.1016/j.xphs.2024.06.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/09/2024]
Abstract
The quantification of both polymer and drug during the dissolution of an amorphous solid dispersion (ASD) in aqueous media arouses great interest and may aid in the formulation. However, the available quantification methods for polymer excipients are limited, expensive, and challenging compared to drugs. In this work, a size exclusion chromatography method (HPLC-SEC) was developed and validated to determine the concentration of a frequently used polymer excipient, Soluplus® (Sol). In order to develop a method for the quantification of dissolved Soluplus®, two methods (SEC-UV and SEC-RID) with two injection volumes were tested with standard solutions of three different batches of Soluplus. The developed HPLC-SEC-UV method showed acceptable linearity (R2 > 0.9990) for all batches of Soluplus, good accuracies above a concentration of 0.1 mg/mL (coefficient of variation < 2 %), relatively good precision at a concentration of 0.1 mg/mL (coefficient of variation < 2.5 %), and high recoveries at a concentration of 0.75 mg/mL (coefficient of variation < 0.5 %). The presence of Felodipine (Fel) and Lumefantrine (Lum) in the liquid media did not interfere with Soluplus quantification. The use of various surfactants, such as Tween® 80, Tween® 20, Span® 80, Span® 20, Kolliphor® TPGS, and sodium lauryl sulphate at a low concentration (0.005 mg/mL) did not show any effect on Soluplus® and did not interfere with Soluplus® quantification with any of the Soluplus batches. The addition of lithium bromide (LiBr) to the mobile phase within a concentration range of 0.05-1.0 M did not improve Soluplus® quantification.
Collapse
Affiliation(s)
- Zoltán Márk Horváth
- Leading Research Group, Faculty of Pharmacy, Rīga Stradiņš University, LV-1007 Riga, Latvia
| | - Liga Petersone
- Leading Research Group, Faculty of Pharmacy, Rīga Stradiņš University, LV-1007 Riga, Latvia.
| | - Valentyn Mohylyuk
- Leading Research Group, Faculty of Pharmacy, Rīga Stradiņš University, LV-1007 Riga, Latvia.
| |
Collapse
|
11
|
Tripathi D, Sharma DK, Sahoo J. Enhancing Ketoprofen Solubility: A Strategic Approach Using Solid Dispersion and Response Surface Methodology. Curr Radiopharm 2025; 18:100-119. [PMID: 39449340 DOI: 10.2174/0118744710311951241018054453] [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: 05/01/2024] [Revised: 08/28/2024] [Accepted: 09/12/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND In the pharmaceutical sciences, the solubility profile of therapeutic molecules is crucial for identifying and formulating drugs and evaluating their quality across the drug discovery pipeline based on factors like oral bioavailability, metabolic transformation, biodistribution kinetics, and potential toxicological implications. The investigation aims to enhance the solubility parameters of ketoprofen (BCS-II class), which exhibits low solubility and high permeability. METHODS In this method, hydrotrope blends of aromatic sodium benzoate and electrolyte sodium acetate were employed to enhance the solubility parameter of ketoprofen. Several batches of solid dispersion of ketoprofen were made using a solvent evaporation method, and the response surface method 3² factorial design was used to find the best one. The optimised formulation, KSD9, underwent in-vitro drug dissolution, DSC, pXRD, and SEM studies. RESULTS The optimized batch demonstrated substantial improvement in ketoprofen solubility, attributed to mixed hydrotropy. The results indicated that both solubility and %CDR improved when hydrotropes were employed, suggesting a direct proportionality between the rise in solubility and % CDR. Formulations KSD1-KSD9 exhibited solubility enhancements ranging from 2.23 to 5.77-fold, along with an elevation in % CDR from 72.28% to 94.76%. This implies that the % CDR was modulated by the hydrotropes, specifically influenced by the concentration levels of the independent variables. An increase in hydrotrope levels corresponded to an increase in % CDR. The positive coefficients in the quadratic equation for % CDR underscored the significant role of these independent variables in augmenting the in-vitro release of Ketoprofen. Similarly, during a comparative dissolution investigation, the optimized KSD9 formulation exhibited remarkable solubility and drug content compared to conventional Ketoprofen dispersible tablets. CONCLUSION The synergistic effect of combining two hydrotropic agents significantly increased the solubility of ketoprofen by up to 58 times. The results indicated that the independent variables exerted a positive influence on solubility and % CDR. Furthermore, the responses were contingent on the specific hydrotropes selected, which functioned as the independent variables. Analyzing the r² and ANOVA results suggested that the dependent variables aligned well with the chosen model. Visual representations, such as the 3D response surface plot and contour plot, demonstrated the impact of each hydrotrope individually and when combined. Overall, employing hydrotropes led to improved solubility and % CDR, highlighting a direct proportionality between the rise in solubility and % CDR. Mixed hydrotropic lessens the toxicity associated with individual hydrotrope concentrations while also offering a sustainable and eco-friendly alternative. This study paves the way for future research aiming to improve the solubility of low- solubility drugs, broadening their clinical applications.
Collapse
Affiliation(s)
- Devika Tripathi
- PSIT-Pranveer Singh Institute of Technology (Pharmacy), Kanpur, Uttar Pradesh, India
- Department of Pharmacy, VMSB, Uttarakhand Technical University, Dehradun, India
| | - Dinesh Kumar Sharma
- School of Pharmacy and Research Centre, Sanskriti University, Mathura, India
| | - Jagannath Sahoo
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, NIMMS, Mumbai, India
| |
Collapse
|
12
|
B S, Ghosh A. Mechanistic Insights into Amorphous Solid Dispersions: Bridging Theory and Practice in Drug Delivery. Pharm Res 2025; 42:1-23. [PMID: 39849216 DOI: 10.1007/s11095-024-03808-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 12/12/2024] [Indexed: 01/25/2025]
Abstract
Improving the bioavailability of poorly water-soluble drugs presents a significant challenge in pharmaceutical development. Amorphous solid dispersions (ASDs) have garnered substantial attention for their capability to augment the solubility and dissolution rate of poorly water-soluble drugs, thereby markedly enhancing their bioavailability. ASDs, characterized by a metastable equilibrium where the active pharmaceutical ingredient (API) is molecularly dispersed, offer enhanced absorption compared to crystalline forms. This review explores recent research advancements in ASD, emphasizing dissolution mechanisms, phase separation phenomena, and the importance of drug loading and congruency limits on ASD performance. Principal occurrences such as liquid-liquid phase separation (LLPS) and supersaturation are discussed, highlighting their impact on drug solubility, absorption and subsequent bioavailability. Additionally, it addresses the role of polymers in controlling supersaturation, stabilizing drug-rich nanodroplets, and inhibiting recrystallization. Recent advancements and emerging technologies offer new avenues for ASD characterization and production and demonstrate the potential of ASDs to enhance bioavailability and reduce variability, making possible for more effective and patient-friendly pharmaceutical formulations. Future research directions are proposed, focusing on advanced computational models for predicting ASD stability, use of novel polymeric carriers, and methods for successful preparations.
Collapse
Affiliation(s)
- Srividya B
- Solid State Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India
| | - Animesh Ghosh
- Solid State Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India.
| |
Collapse
|
13
|
El Sayed M, Alhalaweh A, Kovac L, Bergström CAS. Excipient effects on supersaturation, particle size dynamics, and thermodynamic activity of multidrug amorphous formulations. Int J Pharm 2024; 666:124738. [PMID: 39307444 DOI: 10.1016/j.ijpharm.2024.124738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 10/07/2024]
Abstract
Multidrug formulations enhance patient compliance and extend the life cycle of pharmaceutical products. To overcome solubility challenges for multidrug combinations, amorphous formulations are commonly used. However, the excipients for creating amorphous formulations are often selected without an understanding of their effects on the bioavailability of the drugs. In this context, we investigated the impact of three types of excipients (polymers, surfactants and amino acids) on the supersaturation and thermodynamic activity of multidrug amorphous formulations. Additionally, we studied the particle size dynamics of the colloidal phase formed as a result of liquid-liquid phase separation. The amorphous solubility of two drugs, atazanavir and ritonavir, was determined in solutions containing predissolved excipients and the particle size dynamics of the colloidal particles was measured by dynamic light scattering. Dissolution experiments of atazanavir and ritonavir were conducted in predissolved sodium dodecyl sulfate (SDS), an anionic surfactant, and alanine solutions under non-sink conditions. Membrane transport of the drugs was evaluated using a MicroFLUX setup. The polymers had only minor effects on the amorphous solubility, but SDS significantly increased the solubilities of both drugs. In contrast, the other non-ionic surfactants and amino acids reduced the solubility of atazanavir but had no negative effect on ritonavir. Polymers were effective in maintaining supersaturation and preventing the coarsening of the colloidal particles. Conversely, alanine was neither able to inhibit the solution crystallization nor increase the flux of either drug. Despite the increase in the amorphous solubility of both drugs in SDS, flux was reduced. These results highlight the importance of properly selecting excipients for supersaturating amorphous formulations. The choice of excipient impacts the thermodynamic activity, the phase behaviour of the drugs and hence, the resulting absorption after oral intake.
Collapse
Affiliation(s)
- Mira El Sayed
- Department of Pharmacy, Uppsala University, Biomedical Centre, P.O. Box 580, SE-751 23 Uppsala, Sweden; Recipharm OT Chemistry AB, SE-754 50 Uppsala, Sweden
| | - Amjad Alhalaweh
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Lucia Kovac
- Recipharm OT Chemistry AB, SE-754 50 Uppsala, Sweden
| | - Christel A S Bergström
- Department of Pharmacy, Uppsala University, Biomedical Centre, P.O. Box 580, SE-751 23 Uppsala, Sweden.
| |
Collapse
|
14
|
Polyzois H, Nguyen HT, Roberto de Alvarenga Junior B, Taylor LS. Amorphous Solid Dispersion Formation for Enhanced Release Performance of Racemic and Enantiopure Praziquantel. Mol Pharm 2024; 21:5285-5296. [PMID: 39292641 PMCID: PMC11462518 DOI: 10.1021/acs.molpharmaceut.4c00711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/20/2024]
Abstract
Praziquantel (PZQ) is the treatment of choice for schistosomiasis, which affects more than 250 million people globally. Commercial tablets contain the crystalline racemic compound (RS-PZQ) which limits drug dissolution and oral bioavailability and can lead to unwanted side effects and poor patient compliance due to the presence of the S-enantiomer. While many approaches have been explored for improving PZQ's dissolution and oral bioavailability, studies focusing on investigating its release from amorphous solid dispersions (ASDs) have been limited. In this work, nucleation induction time experiments were performed to identify suitable polymers for preparing ASDs using RS-PZQ and R-PZQ, the therapeutically active enantiomer. Cellulose-based polymers, hydroxypropyl methylcellulose acetate succinate (HPMCAS, MF grade) and hydroxypropyl methylcellulose (HPMC, E5 LV grade), were the best crystallization inhibitors for RS-PZQ in aqueous media and were selected for ASD preparation using solvent evaporation (SE) and hot-melt extrusion (HME). ASDs prepared experimentally were subjected to X-ray powder diffraction to verify their amorphous nature and a selected number of ASDs were monitored and found to remain physically stable following several months of storage under accelerated-stability testing conditions. SE HPMCAS-MF ASDs of RS-PZQ and R-PZQ showed faster release than HPMC E5 LV ASDs and maintained good performance with an increase in drug loading (DL). HME ASDs of RS-PZQ formulated using HPMCAS-MF exhibited slightly enhanced release compared to that of SE ASDs. SE HPMCAS-MF ASDs showed a maximum release increase of the order of 6 times compared to generic and branded (Biltricide) PZQ tablets. More importantly, SE R-PZQ ASDs with HPMCAS-MF released the drug as effectively as RS-PZQ or better, depending on the DL used. These findings have significant implications for the development of commercial PZQ formulations comprised solely of the R-enantiomer, which can result in mitigation of the biopharmaceutical and compliance issues associated with current commercial tablets.
Collapse
Affiliation(s)
- Hector Polyzois
- Department of Industrial and Molecular
Pharmaceutics, College of Pharmacy, Purdue
University, West Lafayette, Indiana 47907, United States
| | - Hanh Thuy Nguyen
- Department of Industrial and Molecular
Pharmaceutics, College of Pharmacy, Purdue
University, West Lafayette, Indiana 47907, United States
| | | | - Lynne S. Taylor
- Department of Industrial and Molecular
Pharmaceutics, College of Pharmacy, Purdue
University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
15
|
Zhang HJ, Chiang CW, Maschmeyer-Tombs T, Conklin B, Napolitano JG, Lubach JW, Nagapudi K, Mao C, Chen Y. Generality of Enhancing the Dissolution Rates of Free Acid Amorphous Solid Dispersions by the Incorporation of Sodium Hydroxide. Mol Pharm 2024; 21:3395-3406. [PMID: 38836777 DOI: 10.1021/acs.molpharmaceut.4c00118] [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: 06/06/2024]
Abstract
The incorporation of a counterion into an amorphous solid dispersion (ASD) has been proven to be an attractive strategy to improve the drug dissolution rate. In this work, the generality of enhancing the dissolution rates of free acid ASDs by incorporating sodium hydroxide (NaOH) was studied by surface-area-normalized dissolution. A set of diverse drug molecules, two common polymer carriers (copovidone or PVPVA and hydroxypropyl methylcellulose acetate succinate or HPMCAS), and two sample preparation methods (rotary evaporation and spray drying) were investigated. When PVPVA was used as the polymer carrier for the drugs in this study, enhancements of dissolution rates from 7 to 78 times were observed by the incorporation of NaOH into the ASDs at a 1:1 molar ratio with respect to the drug. The drugs having lower amorphous solubilities showed greater enhancement ratios, providing a promising path to improve the drug release performance from their ASDs. Samples generated by rotary evaporation and spray drying demonstrated comparable dissolution rates and enhancements when NaOH was added, establishing a theoretical foundation to bridge the ASD dissolution performance for samples prepared by different solvent-removal processes. In the comparison of polymer carriers, when HPMCAS was applied in the selected system (indomethacin ASD), a dissolution rate enhancement of 2.7 times by the incorporated NaOH was observed, significantly lower than the enhancement of 53 times from the PVPVA-based ASD. This was attributed to the combination of a lower dissolution rate of HPMCAS and the competition for NaOH between IMC and HPMCAS. By studying the generality of enhancing ASD dissolution rates by the incorporation of counterions, this study provides valuable insights into further improving drug release from ASD formulations of poorly water-soluble drugs.
Collapse
Affiliation(s)
- Helen J Zhang
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
- Department of Molecular and Cell Biology, University of California, Berkeley, 142 Weill Hall #3200, Berkeley, California 94720, United States
| | - Cheng W Chiang
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Tristan Maschmeyer-Tombs
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Breanna Conklin
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jose G Napolitano
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Joseph W Lubach
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Karthik Nagapudi
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Chen Mao
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Yinshan Chen
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
16
|
Moseson DE, Tran TB, Karunakaran B, Ambardekar R, Hiew TN. Trends in amorphous solid dispersion drug products approved by the U.S. Food and Drug Administration between 2012 and 2023. Int J Pharm X 2024; 7:100259. [PMID: 38974024 PMCID: PMC11225173 DOI: 10.1016/j.ijpx.2024.100259] [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: 05/31/2024] [Accepted: 06/01/2024] [Indexed: 07/09/2024] Open
Abstract
Forty-eight (48) drug products (DPs) containing amorphous solid dispersions (ASDs) have been approved by the U.S. Food and Drug Administration in the 12-year period between 2012 and 2023. These DPs comprise 36 unique amorphous drugs. Ten (10) therapeutic categories are represented, with most DPs containing antiviral and antineoplastic agents. The most common ASD polymers are copovidone (49%) and hypromellose acetate succinate (30%), while spray drying (54%) and hot melt extrusion (35%) are the most utilized manufacturing processes to prepare the ASD drug product intermediate (DPI). Tablet dosage forms are the most common, with several capsule products available. Line extensions of several DPs based on flexible oral solids and powders for oral suspension have been approved which provide patient-centric dosing to pediatric and other patient populations. The trends in the use of common excipients and film coating types are discussed. Eighteen (18) DPs are fixed-dose combinations, and some contain a mixture of amorphous and crystalline drugs. The DPs have dose/unit of amorphous drug ranging from <5 mg up to 300 mg, with the majority being ≤100 mg/unit. This review details several aspects of DPI and DP formulation and manufacturing of ASDs, as well as trends related to therapeutic category, dose, and patient-centricity.
Collapse
Affiliation(s)
- Dana E. Moseson
- Worldwide Research and Development, Pfizer, Inc., Groton, CT 06340, USA
| | - Trong Bien Tran
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa 52242, USA
| | - Bharathi Karunakaran
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa 52242, USA
| | - Rohan Ambardekar
- Worldwide Research and Development, Pfizer, Inc., Sandwich CT13 9NJ, UK
| | - Tze Ning Hiew
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa 52242, USA
| |
Collapse
|
17
|
Petrova SP, Mohamed MA, Wu H, Taylor LS, Edgar KJ. Threading the needle: Achieving simplicity and performance in cellulose alkanoate ω-carboxyalkanoates for amorphous solid dispersion. Carbohydr Polym 2024; 333:121988. [PMID: 38494207 DOI: 10.1016/j.carbpol.2024.121988] [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: 10/31/2023] [Revised: 02/07/2024] [Accepted: 02/24/2024] [Indexed: 03/19/2024]
Abstract
Most active pharmaceutical ingredients (APIs) suffer from poor water solubility, often keeping them from reaching patients. To overcome the issues of poor drug solubility and subsequent low bioavailability, amorphous solid dispersions (ASDs) have garnered much attention. Cellulose ester derivatives are of interest for ASD applications as they are benign, sustainable-based, and successful in commercial drug delivery systems, e.g. in osmotic pump systems and as commercial ASD polymers. Synthesis of carboxy-pendant cellulose esters is a challenge, due in part to competing reactions between carboxyls and hydroxyls, forming ester crosslinks. Herein we demonstrate proof-of-concept for a scalable synthetic route to simple, yet highly promising ASD polymers by esterifying cellulose polymers through ring-opening of cyclic succinic or glutaric anhydride. We describe the complexity of such ring-opening reactions, not previously well-described, and report ways to avoid gelation. We report synthesis, characterization, and preliminary in vitro ASD evaluations of fifteen such derivatives. Synthetic routes were designed to accommodate these criteria: no protecting groups, no metal catalysts, mild conditions with standard reagents, simple purification, and one-pot synthesis. Finally, these designed ASD polymers included members that maintained fast-crystallizing felodipine in solution and release it from an ASD at rather high 20 % drug loading (DL).
Collapse
Affiliation(s)
- Stella P Petrova
- Department of Chemistry, Virginia Tech, Blacksburg, VA 24061, United States of America; Department of Sustainable Biomaterials, Virginia Tech, Blacksburg, VA 24061, United States of America.
| | - Mennatallah A Mohamed
- Department of Industrial and Molecular Pharmaceutics, Purdue University, West Lafayette, IN 47907, United States of America
| | - Huiming Wu
- Department of Chemistry, Virginia Tech, Blacksburg, VA 24061, United States of America
| | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, Purdue University, West Lafayette, IN 47907, United States of America
| | - Kevin J Edgar
- Department of Sustainable Biomaterials, Virginia Tech, Blacksburg, VA 24061, United States of America; Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, United States of America.
| |
Collapse
|
18
|
Jain KMH, Hou HH, Siegel RA. An Artificial Gut/Absorption Simulator: Understanding the Impact of Absorption on In Vitro Dissolution, Speciation, and Precipitation of Amorphous Solid Dispersions. Mol Pharm 2024; 21:1884-1899. [PMID: 38512389 DOI: 10.1021/acs.molpharmaceut.3c01180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Upon dissolution, amorphous solid dispersions (ASDs) of poorly water-soluble compounds can generate supersaturated solutions consisting of bound and free drug species that are in dynamic equilibrium with each other. Only free drug is available for absorption. Drug species bound to bile micelles, polymer excipients, and amorphous and crystalline precipitate can reduce the drug solute's activity to permeate, but they can also serve as reservoirs to replenish free drug in solution lost to absorption. However, with multiple processes of dissolution, absorption, and speciation occurring simultaneously, it may become challenging to understand which processes lead to an increase or decrease in drug solution concentration. Closed, nonsink dissolution testing methods used routinely, in the absence of drug removal, allow only for static equilibrium to exist and obscure the impact of each drug species on absorption. An artificial gut simulator (AGS) introduced recently consists of a hollow fiber-based absorption module and allows mass transfer of the drug from the dissolution media at a physiological rate after tuning the operating parameters. In the present work, ASDs of varying drug loadings were prepared with a BCS-II model compound, ketoconazole (KTZ), and hypromellose acetate succinate (HPMCAS) polymer. Simultaneous dissolution and absorption testing of the ASDs was conducted with the AGS, and simple analytical techniques were utilized to elucidate the impact of bound drug species on absorption. In all cases, a lower amount of crystalline precipitate was formed in the presence of absorption relative to the nonsink dissolution "control". However, formation of HPMCAS-bound drug species and crystalline precipitate significantly reduced KTZ absorption. Moreover, at high drug loading, inclusion of an absorption module was shown to enhance ASD dissolution. The rank ordering of the ASDs with respect to dissolution was significantly different when nonsink dissolution versus AGS was used, and this discrepancy could be mechanistically elucidated by understanding drug dissolution and speciation in the presence of absorption.
Collapse
Affiliation(s)
| | - Hao Helen Hou
- Small Molecule Pharmaceutical Sciences, Genentech Inc., South San Francisco, California 94080, United States
| | - Ronald A Siegel
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
19
|
Deac A, Luebbert C, Qi Q, Courtney RM, Indulkar AS, Gao Y, Zhang GGZ, Sadowski G, Taylor LS. Dissolution Mechanisms of Amorphous Solid Dispersions: Application of Ternary Phase Diagrams To Explain Release Behavior. Mol Pharm 2024; 21:1900-1918. [PMID: 38469754 DOI: 10.1021/acs.molpharmaceut.3c01179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
The use of amorphous solid dispersions (ASDs) in commercial drug products has increased in recent years due to the large number of poorly soluble drugs in the pharmaceutical pipeline. However, the release behavior of ASDs is complex and remains not well understood. Often, the drug release from ASDs is rapid and complete at lower drug loadings (DLs) but becomes slow and incomplete at higher DLs. The DL where release becomes hindered is termed the limit of congruency (LoC). Currently, there are no approaches to predict the LoC. However, recent findings show that one potential cause leading to the LoC is a change in phase morphology after water-induced phase separation at the ASD/solution interface. In this study, the phase behavior of ASDs in contact with aqueous solutions was described thermodynamically by constructing experimental and computational ternary phase diagrams, and these were used to predict morphology changes and ultimately the LoC. Experimental ternary phase diagrams were obtained by equilibrating ASD/water mixtures over time. Computational ternary phase diagrams were obtained by Perturbed Chain Statistical Associating Fluid Theory (PC-SAFT). The morphology of the hydrophobic phase was studied with fluorescence confocal microscopy. It was demonstrated that critical point (plait point) composition approximately corresponded to the ASD DL, where the hydrophobic phase, formed during phase separation, became interconnected and hindered ASD release. This work provides mechanistic insights into the ASD release behavior and highlights the potential of in silico ASD design using phase diagrams.
Collapse
Affiliation(s)
- Alexandru Deac
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | | | - Qingqing Qi
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Reagan M Courtney
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Anura S Indulkar
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Yi Gao
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Geoff G Z Zhang
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | | | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
20
|
Zhuo X, Tozzetti M, Arnous A, Leng D, Foderà V, Löbmann K. Investigating the influence of protein secondary structure on the dissolution behavior of β-lactoglobulin-based amorphous solid dispersions. Int J Pharm 2024; 653:123887. [PMID: 38346599 DOI: 10.1016/j.ijpharm.2024.123887] [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: 01/03/2024] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 02/22/2024]
Abstract
Proteins acting as carriers in amorphous solid dispersions (ASDs) demonstrate a notable sensitivity to the spray drying process, potentially leading to changes in their conformation. The main aim of this study was to investigate the dissolution performance of ASDs based on proteins with different content of secondary structures, specifically β-sheet and α-helix structures. We prepared β-sheet-rich and α-helix-rich β-lactoglobulin (BLG), along with corresponding ASDs containing 10 wt% and 30 wt% drug loadings, through spray drying using celecoxib as the model drug. Circular dichroism and Fourier Transform Infrared Spectroscopy results revealed that even though changes in secondary structure were obtained in the spray-dried powders, the BLGs exhibited reversibility upon re-dissolving in phosphate buffer with varying pH levels. Both β-sheet-rich BLG and α-helix-rich BLG exhibited enhanced dissolution rates and higher solubility in the media with pH values far from the isoelectric point (pI) of BLG (pH 2, 7, 8, and 9) compared to the pH closer to the pI (pH 3, 4, 5, and 6). Notably, the release rate and solubility of the drug and BLG from both types of BLG-based ASDs at 10 wt% drug loading were largely dependent on the solubility of pure SD-BLGs. α-helix-rich BLG-ASDs consistently exhibited equivalent or superior performance to β-sheet-rich BLG-ASDs in terms of drug release rate and solubility, regardless of drug loading. Moreover, both types of BLG-based ASDs at 10 wt% drug loading exhibited faster release rates and higher solubility, for both the drug and BLG, compared to the ASDs at 30 wt% drug loading in pHs 2, 7, and 9 media.
Collapse
Affiliation(s)
- Xuezhi Zhuo
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Martina Tozzetti
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Anis Arnous
- Zerion Pharma A/S, Blokken 11, DK-3460 Birkerød, Denmark
| | - Donglei Leng
- Zerion Pharma A/S, Blokken 11, DK-3460 Birkerød, Denmark
| | - Vito Foderà
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Korbinian Löbmann
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; Zerion Pharma A/S, Blokken 11, DK-3460 Birkerød, Denmark
| |
Collapse
|
21
|
Bapat P, Paul S, Tseng YC, Taylor LS. Interplay of Drug-Polymer Interactions and Release Performance for HPMCAS-Based Amorphous Solid Dispersions. Mol Pharm 2024; 21:1466-1478. [PMID: 38346390 DOI: 10.1021/acs.molpharmaceut.3c01106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
The interplay between drug and polymer chemistry and its impact on drug release from an amorphous solid dispersion (ASD) is a relatively underexplored area. Herein, the release rates of several drugs of diverse chemistry from hydroxypropyl methylcellulose acetate succinate (HPMCAS)-based ASDs were explored using surface area normalized dissolution. The tendency of the drug to form an insoluble complex with HPMCAS was determined through coprecipitation experiments. The role of pH and the extent of drug ionization were probed to evaluate the role of electrostatic interactions in complex formation. Relationships between the extent of complexation and the drug release rate from an ASD were observed, whereby the drugs could be divided into two groups. Drugs with a low extent of insoluble complex formation with HPMCAS tended to be neutral or anionic and showed reasonable release at pH 6.8 even at higher drug loadings. Cationic drugs formed insoluble complexes with HPMCAS and showed poor release when formulated as an ASD. Thus, and somewhat counterintuitively, a weakly basic drug showed a reduced release rate from an ASD at a bulk solution pH where it was ionized, relative to when unionized. The opposite trend was observed in the absence of polymer for the neat amorphous drug. In conclusion, electrostatic interactions between HPMCAS and lipophilic cationic drugs led to insoluble complex formation, which in turn resulted in ASDs with poor release performance.
Collapse
Affiliation(s)
- Pradnya Bapat
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Shubhajit Paul
- Material and Analytical Sciences, Research and Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Yin-Chao Tseng
- Material and Analytical Sciences, Research and Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
22
|
Yang W, Saboo S, Zhou L, Askin S, Bak A. Early evaluation of opportunities in oral delivery of PROTACs to overcome their molecular challenges. Drug Discov Today 2024; 29:103865. [PMID: 38154757 DOI: 10.1016/j.drudis.2023.103865] [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: 10/19/2023] [Revised: 12/08/2023] [Accepted: 12/20/2023] [Indexed: 12/30/2023]
Abstract
PROteolysis TArgeting Chimeras (PROTACs) offer new opportunities in modern medicine by targeting proteins that are intractable to classic inhibitors. Heterobifunctional in nature, PROTACs are small molecules that offer a unique mechanism of protein degradation by hijacking the ubiquitin-mediated protein degradation pathway, known as the ubiquitin-proteasome system. Herein, we present an analysis on the structural characteristics of this novel chemical modality. Furthermore, we review and discuss the formulation opportunities to overcome the oral delivery challenges of PROTACs in drug discovery.
Collapse
Affiliation(s)
- Wenzhan Yang
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Boston, USA.
| | - Sugandha Saboo
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Boston, USA
| | - Liping Zhou
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Boston, USA
| | - Sean Askin
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Annette Bak
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Boston, USA
| |
Collapse
|
23
|
Chiang CW, Tang S, Mao C, Chen Y. Effect of Buffer pH and Concentration on the Dissolution Rates of Sodium Indomethacin-Copovidone and Indomethacin-Copovidone Amorphous Solid Dispersions. Mol Pharm 2023; 20:6451-6462. [PMID: 37917181 DOI: 10.1021/acs.molpharmaceut.3c00827] [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: 11/04/2023]
Abstract
The incorporation of counterions into amorphous solid dispersions (ASDs) has been proven to be effective for improving the dissolution rates of ionizable drugs in ASDs. In this work, the effect of dissolution buffer pH and concentration on the dissolution rate of indomethacin-copovidone 40:60 (IMC-PVPVA, w/w) ASD with or without incorporated sodium hydroxide (NaOH) was studied by surface area-normalized dissolution to provide further mechanistic understanding of this phenomenon. Buffer pH from 4.7 to 7.2 and concentration from 20 to 100 mM at pH 5.5 were investigated. As the buffer pH decreased, the IMC dissolution rate from both ASDs decreased. Compared to IMC-PVPVA ASD, the dissolution rate decrease from IMCNa-PVPVA ASD was more resistant to the decrease of buffer pH. In contrast, while buffer concentration had a negligible impact on the IMC dissolution rate from IMC-PVPVA ASD, the increase of buffer concentration significantly reduced the IMC dissolution rate from IMCNa-PVPVA ASD. Surrogate evaluation of microenvironment pH modification by the dissolution of IMCNa-PVPVA ASD demonstrated the successful elevation of buffer microenvironment pH and the suppression of such pH elevation by the increase of buffer concentration. These results are consistent with the hypothesis that the dissolution rate enhancement by the incorporation of counterions originates from the enhanced drug solubility by ionization and the modification of diffusion layer pH in favor of drug dissolution. At the studied drug loading (∼40%), relatively congruent release between IMC and PVPVA was observed when IMC was ionized in ASD or in solution, highlighting the importance of studying the ionization effect on the congruent release of ASDs, especially when drug ionization is expected in vivo. Overall, this work further supports the application of incorporating counterions into ASDs for improving the dissolution rates of ionizable drugs when enabling formulation development is needed.
Collapse
Affiliation(s)
- Cheng W Chiang
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Shijia Tang
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Chen Mao
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Yinshan Chen
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
24
|
Bapat P, Paul S, Thakral NK, Tseng YC, Taylor LS. Does Media Choice Matter When Evaluating the Performance of Hydroxypropyl Methylcellulose Acetate Succinate-Based Amorphous Solid Dispersions? Mol Pharm 2023; 20:5714-5727. [PMID: 37751517 DOI: 10.1021/acs.molpharmaceut.3c00586] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
Hydroxypropyl methylcellulose acetate succinate (HPMCAS) is a weakly acidic polymer that is widely used in the formulation of amorphous solid dispersions (ASDs). While the pH-dependent solubility of HPMCAS is widely recognized, the role of other solution properties, including buffer capacity, is less well understood in the context of ASD dissolution. The goal of this study was to elucidate the rate-limiting steps for drug and HPMCAS release from ASDs formulated with two poorly water soluble model drugs, indomethacin and indomethacin methyl ester. The surface area normalized release rate of the drug and/or polymer in a variety of media was determined. The HPMCAS gel layer apparent pH was determined by incorporating pH sensitive dyes into the polymer matrix. Water uptake extent and rate into the ASDs were measured gravimetrically. For neat HPMCAS, the rate-limiting step for polymer dissolution was observed to be the polymer solubility at the polymer-solution interface. This, in turn, was impacted by the gel layer pH which was found to be substantially lower than the bulk solution pH, varying with medium buffer capacity. For the ASDs, the HPMCAS release rate was found to control the drug release rate. However, both drugs reduced the polymer release rate with indomethacin methyl ester having a larger impact. In low buffer capacity media, the presence of the drug had less impact on release rates when compared to observations in higher strength buffers, suggesting changes in the rate-limiting steps for HPMCAS dissolution. The observations made in this study can contribute to the fundamental understanding of acidic polymer dissolution in the presence and absence of a molecularly dispersed lipophilic drug and will help aid in the design of more in vivo relevant release testing experiments.
Collapse
Affiliation(s)
- Pradnya Bapat
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Shubhajit Paul
- Material and Analytical Sciences, Research and Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Naveen K Thakral
- Material and Analytical Sciences, Research and Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Yin-Chao Tseng
- Material and Analytical Sciences, Research and Development, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
25
|
Bharti K, Deepika D, Kumar M, Jha A, Manjit, Akhilesh, Tiwari V, Kumar V, Mishra B. Development and Evaluation of Amorphous Solid Dispersion of Riluzole with PBPK Model to Simulate the Pharmacokinetic Profile. AAPS PharmSciTech 2023; 24:219. [PMID: 37891363 DOI: 10.1208/s12249-023-02680-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
In the current work, screening of polymers viz. polyacrylic acid (PAA), polyvinyl pyrrolidone vinyl acetate (PVP VA), and hydroxypropyl methyl cellulose acetate succinate (HPMC AS) based on drug-polymer interaction and wetting property was done for the production of a stable amorphous solid dispersion (ASD) of a poorly water-soluble drug Riluzole (RLZ). PAA showed maximum interaction and wetting property hence, was selected for further studies. Solid state characterization studies confirmed the formation of ASD with PAA. Saturation solubility, dissolution profile, and in vivo pharmacokinetic data of the ASD formulation were generated in rats against its marketed tablet Rilutor. The RLZ:PAA ASD showed exponential enhancement in the dissolution of RLZ. Predicted and observed pharmacokinetic data in rats showed enhanced area under curve (AUC) and Cmax in plasma and brain with respect to Rilutor. Furthermore, a physiologically based pharmacokinetic (PBPK) model of rats for Rilutor and RLZ ASD was developed and then extrapolated to humans where physiological parameters were changed along with a biochemical parameter. The partition coefficient was kept similar in both species. The model was used to predict different exposure scenarios, and the simulated data was compared with observed data points. The PBPK model simulated Cmax and AUC was within two times the experimental data for plasma and brain. The Cmax and AUC in the brain increased with ASD compared to Rilutor for humans showing its potential in improving its biopharmaceutical performance and hence enhanced therapeutic efficacy. The model can predict the RLZ concentration in multiple compartments including plasma and liver.
Collapse
Affiliation(s)
- Kanchan Bharti
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Deepika Deepika
- Environmental Engineering Laboratory, Departament d' Enginyeria Quimica, Universitat Rovira i Virgili, Tarragona, Catalonia, Spain
- Pere Virgili Health Research Institute (IISPV), Hospital Universitari Sant Joan de Reus, Universitat Rovira I Virgili, Reus, Catalonia, Spain
| | - Manish Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Abhishek Jha
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Manjit
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Akhilesh
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Vinod Tiwari
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Vikas Kumar
- Environmental Engineering Laboratory, Departament d' Enginyeria Quimica, Universitat Rovira i Virgili, Tarragona, Catalonia, Spain
- Pere Virgili Health Research Institute (IISPV), Hospital Universitari Sant Joan de Reus, Universitat Rovira I Virgili, Reus, Catalonia, Spain
- German Federal Institute for Risk Assessment (BfR), Department of Pesticides Safety, Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Brahmeshwar Mishra
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India.
| |
Collapse
|
26
|
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
|
27
|
Bertoni S, Albertini B, Ronowicz-Pilarczyk J, Passerini N. Tailoring the release of drugs having different water solubility by hybrid polymer-lipid microparticles with a biphasic structure. Eur J Pharm Biopharm 2023; 190:171-183. [PMID: 37517450 DOI: 10.1016/j.ejpb.2023.07.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/25/2023] [Accepted: 07/28/2023] [Indexed: 08/01/2023]
Abstract
The aim of this study is to investigate the potential of hybrid polymer-lipid microparticles with a biphasic structure (b-MPs) as drug delivery system. Hybrid b-MPs of Compritol®888 ATO as main lipid constituent of the shell and polyethylene glycol 400 as core material were produced by an innovative solvent-free approach based on spray congealing. To assess the suitability of hybrid b-MPs to encapsulate various types of APIs, three model drugs (fluconazole, tolbutamide and nimesulide) with extremely different water solubility were loaded into the polymeric core. The hybrid systems were characterized in terms of particle size, morphology and physical state. Various techniques (e.g. optical, Confocal Raman and Scanning Electron Microscopy) were used to investigate the influence of the drugs on different aspects of the b-MPs, including external and internal morphology, properties at the lipid/polymer interface and drug distribution. Hybrid b-MPs were suitable for the encapsulation of all drugs (encapsulation efficiency > 90 %) regardless the drug hydrophobic/hydrophilic properties. Finally, the drug release behaviors from hybrid b-MPs were studied and compared with traditional solid lipid MPs (consisting of only the lipid carrier). Due to the combination of lipid and polymeric materials, hybrid b-MPs showed a wide array of release profiles that depends on their composition, the type of loaded drug, the drug loading amount and location, providing a versatile platform and allowing the formulators to finely balance the release performance of drugs intended for oral administration. Overall, the study demonstrates that hybrid, solvent-free b-MPs produced by spray congealing are an extremely versatile delivery platform able to efficiently encapsulate and release very different types of drug compounds.
Collapse
Affiliation(s)
- Serena Bertoni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via S. Donato 19/2, Bologna 40127, Italy
| | - Beatrice Albertini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via S. Donato 19/2, Bologna 40127, Italy.
| | - Joanna Ronowicz-Pilarczyk
- Department of Inorganic and Analytical Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Jurasza 2, Bydgoszcz 85-089, Poland
| | - Nadia Passerini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via S. Donato 19/2, Bologna 40127, Italy
| |
Collapse
|
28
|
Li J, Wang Y, Yu D. Effects of Additives on the Physical Stability and Dissolution of Polymeric Amorphous Solid Dispersions: a Review. AAPS PharmSciTech 2023; 24:175. [PMID: 37603110 DOI: 10.1208/s12249-023-02622-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
Polymeric amorphous solid dispersion (ASD) is a popular approach for enhancing the solubility of poorly water-soluble drugs. However, achieving both physical stability and dissolution performance in an ASD prepared with a single polymer can be challenging. Therefore, a secondary excipient can be added. In this paper, we review three classes of additives that can be added internally to ASDs: (i) a second polymer, to form a ternary drug-polymer-polymer ASD, (ii) counterions, to facilitate in situ salt formation, and (iii) surfactants. In an ASD prepared with a combination of polymers, each polymer exerts a unique function, such as a stabilizer in the solid state and a crystallization inhibitor during dissolution. In situ salt formation in ASD usually leads to substantial increases in the glass transition temperature, contributing to improved physical stability. Surfactants can enhance the wettability of ASD particles, thereby promoting rapid drug release. However, their potential adverse effects on physical stability and dissolution, resulting from enhanced molecular mobility and competitive molecular interaction with the polymer, respectively, warrant careful consideration. Finally, we discuss the impact of magnesium stearate and inorganic salts, excipients added externally upon downstream processing, on the solid-state stability as well as the dissolution of ASD tablets.
Collapse
Affiliation(s)
- Jinghan Li
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, 55455, USA
| | - Yihan Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, 20 North Pine Street, Baltimore, Maryland, 21201, USA
| | - Dongyue Yu
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, New Jersey, 08540, USA.
| |
Collapse
|
29
|
Zhang J, Guo M, Luo M, Cai T. Advances in the development of amorphous solid dispersions: The role of polymeric carriers. Asian J Pharm Sci 2023; 18:100834. [PMID: 37635801 PMCID: PMC10450425 DOI: 10.1016/j.ajps.2023.100834] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/26/2023] [Accepted: 07/23/2023] [Indexed: 08/27/2023] Open
Abstract
Amorphous solid dispersion (ASD) is one of the most effective approaches for delivering poorly soluble drugs. In ASDs, polymeric materials serve as the carriers in which the drugs are dispersed at the molecular level. To prepare the solid dispersions, there are many polymers with various physicochemical and thermochemical characteristics available for use in ASD formulations. Polymer selection is of great importance because it influences the stability, solubility and dissolution rates, manufacturing process, and bioavailability of the ASD. This review article provides a comprehensive overview of ASDs from the perspectives of physicochemical characteristics of polymers, formulation designs and preparation methods. Furthermore, considerations of safety and regulatory requirements along with the studies recommended for characterizing and evaluating polymeric carriers are briefly discussed.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China
| | - Minshan Guo
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Minqian Luo
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Ting Cai
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
30
|
Thompson SA, Gala U, Davis DA, Kucera S, Miller D, Williams RO. Can the Oral Bioavailability of the Discontinued Prostate Cancer Drug Galeterone Be Improved by Processing Method? KinetiSol® Outperforms Spray Drying in a Head-to-head Comparison. AAPS PharmSciTech 2023; 24:137. [PMID: 37344629 DOI: 10.1208/s12249-023-02597-6] [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: 03/28/2023] [Accepted: 05/31/2023] [Indexed: 06/23/2023] Open
Abstract
Galeterone, a novel prostate cancer candidate treatment, was discontinued after a Phase III clinical trial due to lack of efficacy. Galeterone is weakly basic and exhibits low solubility in biorelevant media (i.e., ~ 2 µg/mL in fasted simulated intestinal fluid). It was formulated as a 50-50 (w/w) galeterone-hypromellose acetate succinate spray-dried dispersion to increase its bioavailability. Despite this increase, the bioavailability of this formulation may have been insufficient and contributed to its clinical failure. We hypothesized that reformulating galeterone as an amorphous solid dispersion by KinetiSol® compounding could increase its bioavailability. In this study, we examined the effects of composition and manufacturing technology (Kinetisol and spray drying) on the performance of galeterone amorphous solid dispersions. KinetiSol compounding was utilized to create galeterone amorphous solid dispersions containing the complexing agent hydroxypropyl-β-cyclodextrin or hypromellose acetate succinate with lower drug loads that both achieved a ~ 6 × increase in dissolution performance versus the 50-50 spray-dried dispersion. When compared to a spray-dried dispersion with an equivalent drug load, the KinetiSol amorphous solid dispersions formulations exhibited ~ 2 × exposure in an in vivo rat study. Acid-base surface energy analysis showed that the equivalent composition of the KinetiSol amorphous solid dispersion formulation better protected the weakly basic galeterone from premature dissolution in acidic media and thereby reduced precipitation, inhibited recrystallization, and extended the extent of supersaturation during transit into neutral intestinal media.
Collapse
Affiliation(s)
- Stephen A Thompson
- Molecular Pharmaceutics and Drug Delivery Division, The University of Texas at Austin College of Pharmacy, 2409 W. University Ave. PHR 4.214, Austin, Texas, 78712, USA.
| | - Urvi Gala
- AustinPx, LLC. 111 W Cooperative Way, Suite 300, Georgetown, Texas, 78626, USA
| | - Daniel A Davis
- AustinPx, LLC. 111 W Cooperative Way, Suite 300, Georgetown, Texas, 78626, USA
| | - Sandra Kucera
- AustinPx, LLC. 111 W Cooperative Way, Suite 300, Georgetown, Texas, 78626, USA
| | - Dave Miller
- AustinPx, LLC. 111 W Cooperative Way, Suite 300, Georgetown, Texas, 78626, USA
| | - Robert O Williams
- Molecular Pharmaceutics and Drug Delivery Division, The University of Texas at Austin College of Pharmacy, 2409 W. University Ave. PHR 4.214, Austin, Texas, 78712, USA
| |
Collapse
|
31
|
Nyamba I, Jennotte O, Sombie CB, Lechanteur A, Sacre PY, Djande A, Semde R, Evrard B. Preformulation study for the selection of a suitable polymer for the development of ellagic acid-based solid dispersion using hot-melt extrusion. Int J Pharm 2023:123088. [PMID: 37257795 DOI: 10.1016/j.ijpharm.2023.123088] [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: 10/10/2022] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/02/2023]
Abstract
Ellagic acid is one of the most studied polyphenolic compounds due to its numerous promising therapeutic properties. However, this therapeutic potential remains difficult to exploit owing to its low solubility and low permeability, resulting in low oral bioavailability. In order to allow an effective therapeutic application of EA, it is therefore necessary to develop strategies that sufficiently enhance its solubility, dissolution rate and bioavailability. For this purpose, solid dispersions based on pre-selected polymers such as Eudragit® EPO, Soluplus® and Kollidon® VA 64, with 5% w/w ellagic acid loading were prepared by hot extrusion and characterized by X-ray diffraction, FTIR spectroscopy and in vitro dissolution tests in order to select the most suitable polymer for future investigations. The results showed that Eudragit® EPO was the most promising polymer for ellagic acid solid dispersions development because its extrudates allowed to obtain a solution supersaturated in ellagic acid that was stable for at least 90 min. Moreover, the resulting apparent solubility was 20 times higher than the actual solubility of ellagic acid. The extrudates also showed a high dissolution rate of ellagic acid (96.25% in 15 min), compared to the corresponding physical mixture (6.52% in 15 min) or the pure drug (1.56% in 15 min). Furthermore, increasing the loading rate of ellagic acid up to 12% in extrudates based on this polymer did not negatively influence its release profile through dissolution tests.
Collapse
Affiliation(s)
- Isaïe Nyamba
- Laboratory of Pharmaceutical Technology and Biopharmacy, Center for Interdisciplinary Research on Medicines (CIRM), Université de Liège, 4000 Liège (Belgium); Laboratory of Drug Development, Center of training, research and expertise in pharmaceutical sciences (CFOREM), Doctoral School of Sciences and Health, Université Joseph KI-ZERBO, 03 BP 7021 Ouagadougou 03 (Burkina Faso).
| | - Olivier Jennotte
- Laboratory of Pharmaceutical Technology and Biopharmacy, Center for Interdisciplinary Research on Medicines (CIRM), Université de Liège, 4000 Liège (Belgium)
| | - Charles B Sombie
- Laboratory of Drug Development, Center of training, research and expertise in pharmaceutical sciences (CFOREM), Doctoral School of Sciences and Health, Université Joseph KI-ZERBO, 03 BP 7021 Ouagadougou 03 (Burkina Faso)
| | - Anna Lechanteur
- Laboratory of Pharmaceutical Technology and Biopharmacy, Center for Interdisciplinary Research on Medicines (CIRM), Université de Liège, 4000 Liège (Belgium)
| | - Pierre-Yves Sacre
- Laboratory of Pharmaceutical Analytical Chemistry, Department of Pharmacy, Center for Interdisciplinary Research on Medicines (CIRM), University of Liege, 4000 Liege, Belgium
| | - Abdoulaye Djande
- Department of Chemistry, Laboratory of Molecular Chemistry and Materials, Research Team: Organic Chemistry and Phytochemistry, Université Joseph KI-ZERBO, 03 BP 7021 Ouagadougou 03 (Burkina Faso)
| | - Rasmané Semde
- Laboratory of Drug Development, Center of training, research and expertise in pharmaceutical sciences (CFOREM), Doctoral School of Sciences and Health, Université Joseph KI-ZERBO, 03 BP 7021 Ouagadougou 03 (Burkina Faso)
| | - Brigitte Evrard
- Laboratory of Pharmaceutical Technology and Biopharmacy, Center for Interdisciplinary Research on Medicines (CIRM), Université de Liège, 4000 Liège (Belgium)
| |
Collapse
|
32
|
Thompson SA, Davis DA, Miller DA, Kucera SU, Williams RO. Pre-Processing a Polymer Blend into a Polymer Alloy by KinetiSol Enables Increased Ivacaftor Amorphous Solid Dispersion Drug Loading and Dissolution. Biomedicines 2023; 11:1281. [PMID: 37238952 PMCID: PMC10215938 DOI: 10.3390/biomedicines11051281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/13/2023] [Accepted: 04/20/2023] [Indexed: 05/28/2023] Open
Abstract
This study compares the effects of pre-processing multiple polymers together to form a single-phase polymer alloy prior to amorphous solid dispersion formulation. KinetiSol compounding was used to pre-process a 1:1 (w/w) ratio of hypromellose acetate succinate and povidone to form a single-phase polymer alloy with unique properties. Ivacaftor amorphous solid dispersions comprising either a polymer, an unprocessed polymer blend, or the polymer alloy were processed by KinetiSol and examined for amorphicity, dissolution performance, physical stability, and molecular interactions. A polymer alloy ivacaftor solid dispersion with a drug loading of 50% w/w was feasible versus 40% for the other compositions. Dissolution in fasted simulated intestinal fluid revealed that the 40% ivacaftor polymer alloy solid dispersion reached a concentration of 595 µg/mL after 6 h, 33% greater than the equivalent polymer blend dispersion. Fourier transform infrared spectroscopy and solid-state nuclear magnetic resonance revealed changes in the ability of the povidone contained in the polymer alloy to hydrogen bond with the ivacaftor phenolic moiety, explaining the differences in the dissolution performance. This work demonstrates that the creation of polymer alloys from polymer blends is a promising technique that provides the ability to tailor properties of a polymer alloy to maximize the drug loading, dissolution performance, and stability of an ASD.
Collapse
Affiliation(s)
- Stephen A. Thompson
- Molecular Pharmaceutics and Drug Delivery Division, College of Pharmacy, The University of Texas at Austin, 2409 W. University Ave, PHR 4.214, Austin, TX 78712, USA
| | - Daniel A. Davis
- AustinPx, LLC, 111 W Cooperative Way, Suite 300, Georgetown, TX 78626, USA
| | - Dave A. Miller
- AustinPx, LLC, 111 W Cooperative Way, Suite 300, Georgetown, TX 78626, USA
| | - Sandra U. Kucera
- AustinPx, LLC, 111 W Cooperative Way, Suite 300, Georgetown, TX 78626, USA
| | - Robert O. Williams
- Molecular Pharmaceutics and Drug Delivery Division, College of Pharmacy, The University of Texas at Austin, 2409 W. University Ave, PHR 4.214, Austin, TX 78712, USA
| |
Collapse
|
33
|
Deac A, Qi Q, Indulkar AS, Gao Y, Zhang GGZ, Taylor LS. Dissolution Mechanisms of Amorphous Solid Dispersions: A Close Look at the Dissolution Interface. Mol Pharm 2023; 20:2217-2234. [PMID: 36926898 DOI: 10.1021/acs.molpharmaceut.3c00020] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Despite the recent success of amorphous solid dispersions (ASDs) at enabling the delivery of poorly soluble small molecule drugs, ASD-based dosage forms are limited by low drug loading. This is partially due to a sharp decline in drug release from the ASD at drug loadings surpassing the 'limit of congruency' (LoC). In some cases, the LoC is as low as 5% drug loading, significantly increasing the risk of pill burden. Despite efforts to understand the mechanism responsible for the LoC, a clear picture of the molecular processes occurring at the ASD/solution interface remains elusive. In this study, the ASD/solution interface was studied for two model compounds formulated as ASDs with copovidone. The evolution of a gel layer and its phase behavior was captured in situ with fluorescence confocal microscopy, where fluorescent probes were added to label the hydrophobic and hydrophilic phases. Phase separation was detected in the gel layer for most of the ASDs. The morphology of the hydrophobic phase was found to correlate with the release behavior, where a discrete phase resulted in good release and a continuous phase formed a barrier leading to poor release. The continuous phase formed at a lower drug loading for the system with stronger drug-polymer interactions. This was due to incorporation of the polymer into the hydrophobic phase. The study highlights the complex molecular and phase behavior at the ASD/solution interface of copovidone-based ASDs and provides a thermodynamic argument for qualitatively predicting the release behavior based on drug-polymer interactions.
Collapse
Affiliation(s)
- Alexandru Deac
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Qingqing Qi
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Anura S Indulkar
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Yi Gao
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Geoff G Z Zhang
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
34
|
Leng D, Bulduk B, Anlahr J, Müllers W, Löbmann K. Enhanced dissolution rate of nimodipine through β-lactoglobulin based formulation. Int J Pharm 2023; 635:122693. [PMID: 36754186 DOI: 10.1016/j.ijpharm.2023.122693] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/19/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023]
Abstract
Amorphous solid dispersions (ASD) have been considered as one of the most effective strategies to increase solubility and dissolution rate of poorly water-soluble drugs. Carriers, in which the poorly water-soluble drug is dispersed, contribute a large extent to the solid-state properties, stabilities and dissolution performance of ASDs. This study investigated the solid-state properties, physical stability, and in vitro dissolution behaviour of nimodipine ASDs formulated with a traditional polymeric carrier, i.e., polyvinylpyrrolidone (PVP) and a novel carrier, i.e., β-lactoglobulin (BLG). The ASDs with both carriers were prepared using ball milling as preparative technique at 10 %, 17.5 %, 25 %, 30 % and 40 % drug loadings (DLs). All the formulations were found to be amorphous upon milling for 60 min based on X-ray powder diffraction measurements, however, the ASDs were found to be homogeneous unequivocally only at DLs below 25 %. After open storage at accelerated conditions (40 °C/75 % relative humidity), only the ASDs formulated with BLG at 10 % and 17.5 % DLs maintained the amorphous form. The dissolution study revealed that all the freshly prepared ASDs formulated with PVP and the ASDs formulated with BLG at or above 25 % DLs, showed a low drug release (<30 µg/mL in simulated gastric fluid, < 70 µg/mL in simulated intestinal fluid). Whilst the ASD formulated with BLG at 10 % DL exhibited a high drug release with a maximum concentration (Cmax) of 251 µg/mL in simulated gastric fluid and 231 µg/mL in simulated intestinal fluid. Surprisingly, the ASD formulated with BLG at 17.5 % DL demonstrated an even higher drug release (Cmax, 643 µg/mL in simulated gastric fluid, 332 µg/mL in simulated intestinal fluid), compared to the ASD of 10 % DL. These findings underline the importance of rationally investigating both carrier types and DL in the design of ASDs, in order to obtain a stable ASD with the desired enhanced dissolution rate of poorly water-soluble drugs.
Collapse
Affiliation(s)
| | | | - Johanna Anlahr
- Bayer AG, Research and Development, Pharmaceuticals, Wuppertal, Germany
| | - Wouter Müllers
- Bayer AG, Research and Development, Pharmaceuticals, Berlin, Germany
| | | |
Collapse
|
35
|
Yao X, Yu L, Zhang GGZ. Impact of Crystal Nuclei on Dissolution of Amorphous Drugs. Mol Pharm 2023; 20:1796-1805. [PMID: 36749110 DOI: 10.1021/acs.molpharmaceut.2c00989] [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: 02/08/2023]
Abstract
Amorphous drugs are used to improve bioavailability of poorly water-soluble drugs. Crystallization must be managed to take full advantage of this formulation strategy. Crystallization of amorphous drugs proceeds in a sequence of crystal nucleation and growth, with different kinetics. At low temperatures, crystal nucleation is fast, but crystal growth is slow. Therefore, amorphous drugs may generate dense but nanoscale crystal nuclei. Such tiny nuclei cannot be detected using routine powder X-ray diffraction (PXRD) and polarized light microscopy (PLM). However, they may negate the dissolution advantage of amorphous drugs. In this work, for the first time, the impact of crystal nuclei on dissolution of amorphous drugs was studied by monitoring the real-time dissolution from amorphous drug films, with and without crystal nuclei, and the evolving crystallinity in the films. Three model drugs (ritonavir/RTV, posaconazole/POS, and nifedipine/NIF) were chosen to represent different crystallization tendencies in the supercooled liquid state, namely, slow-nucleation-and-slow-growth (SN-SG), fast-nucleation-and-slow-growth (FN-SG), and fast-nucleation-and-fast-growth (FN-FG), respectively. We find that although the amorphous films containing nuclei do not show obvious differences from the nuclei-free films under PLM and PXRD before dissolution, they have inferior dissolution performance relative to the nuclei-free amorphous films. For SN-SG drug RTV, crystal nuclei have negligible impact on the crystallization of amorphous films, dissolution rate, and supersaturation achieved. However, they cause earlier de-supersaturation by inducing crystallization in solution as heterogeneous seeds. For FN-SG drug POS and FN-FG drug NIF, crystal nuclei accelerate crystallization in the amorphous films leading to lower supersaturation achieved with POS, and elimination of any supersaturation with NIF. Dissolution profiles of amorphous films can be further analyzed using a derivative function of the apparent dissolution rate, which yields amorphous solubility, initial intrinsic dissolution rate, and onset of crystallization in the amorphous films. This study highlights that although crystal nuclei are undetectable with routine analytical methods, they can significantly negate, or even eliminate, the dissolution advantage of amorphous drugs. Hence, understanding crystal nucleation process and developing approaches to prevent it are necessary to fully realize the benefits of amorphous solids.
Collapse
Affiliation(s)
| | | | - Geoff G Z Zhang
- Development Sciences, Research and Development, AbbVie, Inc., North Chicago, Illinois 60064, United States
| |
Collapse
|
36
|
Correa-Soto CE, Gao Y, Indulkar AS, Zhang GGZ, Taylor LS. Release Enhancement by Plasticizer Inclusion for Amorphous Solid Dispersions Containing High T g Drugs. Pharm Res 2023; 40:777-790. [PMID: 36859747 DOI: 10.1007/s11095-023-03483-3] [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: 12/13/2022] [Accepted: 02/10/2023] [Indexed: 03/03/2023]
Abstract
PURPOSE Plasticizers are commonly used in the preparation of amorphous solid dispersions (ASDs) with the main goal of aiding processability; however, to the best of our knowledge, the impact of plasticizers on drug release has not been explored. The goal of this study was to evaluate diverse plasticizers, including glycerol and citrate derivatives, as additives to increase the drug loading where good drug release could be achieved from copovidone (PVPVA)-based dispersions, focusing on high glass transition (Tg) drugs, atazanavir (ATZ) and ledipasvir (LED). METHODS ASDs were prepared using the high Tg compounds, atazanavir (ATZ) and ledipasvir (LED), as model drugs. Release was evaluated using surface normalized dissolution testing. Differential scanning calorimetry was used to measure glass transition temperature and water vapor sorption was performed on select samples. RESULTS The presence of a plasticizer at 5% w/w for ATZ and 10% w/w for LED ASDs, led to improved drug release. For ATZ ASDs, in the absence of plasticizer, release was very poor at drug loadings of 10% w/w and above. Good release was obtained for plasticized ASDs up to a drug loading of 25%. The corresponding improvement for LED was from 5 to 20% DL. Interestingly, for a low Tg compound, ritonavir, relatively smaller improvements in release as a function of drug loading were achieved through plasticizer incorporation. CONCLUSIONS The use of plasticizers represents a potential new strategy to increase drug loading in ASDs for high Tg compounds with a low tendency to crystallize and may help improve a major limitation of ASD formulations, namely the high excipient burden.
Collapse
Affiliation(s)
- Clara E Correa-Soto
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
- Pivotal Drug Product, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, USA
| | - Yi Gao
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, IL, 60064, USA
| | - Anura S Indulkar
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, IL, 60064, USA
| | - Geoff G Z Zhang
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, IL, 60064, USA.
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
37
|
Jørgensen JR, Mohr W, Rischer M, Sauer A, Mistry S, Müllertz A, Rades T. Stability and intrinsic dissolution of vacuum compression molded amorphous solid dispersions of efavirenz. Int J Pharm 2023; 632:122564. [PMID: 36586638 DOI: 10.1016/j.ijpharm.2022.122564] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/23/2022] [Accepted: 12/26/2022] [Indexed: 12/29/2022]
Abstract
In this study, the stability and intrinsic dissolution of vacuum compression molded (VCM) amorphous solid dispersions (ASDs) of efavirenz (EFV) were investigated in relation to its solubility limits in seven polymers determined by the melting point depression (MPD) method. The extrapolated solubility limits of EFV at 22 °C ranged from 3 to 68% (w/w) with PVOH being the only polymer suggesting immiscibility with EFV according to both MPD and Hansen solubility parameters (HSPs). All ASDs with EFV loadings below or close to their calculated solubility limit did not show any signs of crystallization upon conditioning for 7 months at either 22 or 37 °C and 23 or 75% relative humidity. However, all ASDs with EFV loading above the solubility limit crystallized at high humidity, while the ASDs with cellulose derived carrier polymers proved kinetically stable at low humidity over 7 months. While the EFV intrinsic dissolution rates from the VCM ASDs were partly depending on the polymer dissolution rate, no correlation was observed between EFV matrix crystallization and its miscibility in the polymer. Altogether, the observations of the study underline the importance of combining preformulation miscibility determination and dissolution studies to rationally decide on both stability and viability of ASD formulations.
Collapse
Affiliation(s)
- Jacob Rune Jørgensen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Wolfgang Mohr
- Losan Pharma GmbH, Otto-Hahn-Str. 13, 79395 Neuenburg, Germany
| | | | - Andreas Sauer
- SE Tylose GmbH & Co. KG, Kasteler Str. 45, 65203 Wiesbaden, Germany
| | - Shilpa Mistry
- Harke Pharma GmbH, Xantener Str. 1, 45479 Mülheim a. d. Ruhr, Germany
| | - Anette Müllertz
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; Bioneer:FARMA, Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| | - Thomas Rades
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| |
Collapse
|
38
|
Cao Z, Harmon DM, Yang R, Razumtcev A, Li M, Carlsen MS, Geiger AC, Zemlyanov D, Sherman AM, Takanti N, Rong J, Hwang Y, Taylor LS, Simpson GJ. Periodic Photobleaching with Structured Illumination for Diffusion Imaging. Anal Chem 2023; 95:2192-2202. [PMID: 36656303 DOI: 10.1021/acs.analchem.2c02950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The use of periodically structured illumination coupled with spatial Fourier-transform fluorescence recovery after photobleaching (FT-FRAP) was shown to support diffusivity mapping within segmented domains of arbitrary shape. Periodic "comb-bleach" patterning of the excitation beam during photobleaching encoded spatial maps of diffusion onto harmonic peaks in the spatial Fourier transform. Diffusion manifests as a simple exponential decay of a given harmonic, improving the signal to noise ratio and simplifying mathematical analysis. Image segmentation prior to Fourier transformation was shown to support pooling for signal to noise enhancement for regions of arbitrary shape expected to exhibit similar diffusivity within a domain. Following proof-of-concept analyses based on simulations with known ground-truth maps, diffusion imaging by FT-FRAP was used to map spatially-resolved diffusion differences within phase-separated domains of model amorphous solid dispersion spin-cast thin films. Notably, multi-harmonic analysis by FT-FRAP was able to definitively discriminate and quantify the roles of internal diffusion and exchange to higher mobility interfacial layers in modeling the recovery kinetics within thin amorphous/amorphous phase-separated domains, with interfacial diffusion playing a critical role in recovery. These results have direct implications for the design of amorphous systems for stable storage and efficacious delivery of therapeutic molecules.
Collapse
Affiliation(s)
- Ziyi Cao
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana47907, United States
| | - Dustin M Harmon
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana47907, United States
| | - Ruochen Yang
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana47907, United States
| | - Aleksandr Razumtcev
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana47907, United States
| | - Minghe Li
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana47907, United States
| | - Mark S Carlsen
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana47907, United States
| | - Andreas C Geiger
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana47907, United States
| | - Dmitry Zemlyanov
- Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana47907, United States
| | - Alex M Sherman
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana47907, United States
| | - Nita Takanti
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana47907, United States
| | - Jiayue Rong
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana47907, United States
| | - Yechan Hwang
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana47907, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana47907, United States
| | - Garth J Simpson
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana47907, United States
| |
Collapse
|
39
|
Supersaturation and phase behavior during dissolution of amorphous solid dispersions. Int J Pharm 2023; 631:122524. [PMID: 36549404 DOI: 10.1016/j.ijpharm.2022.122524] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/04/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022]
Abstract
Amorphous solid dispersion (ASD) is a promising strategy to enhance solubility and bioavailability of poorly water-soluble drugs. Due to higher free energy of ASD, supersaturated drug solution could be generated during dissolution. When amorphous solubility of a drug is exceeded, drug-rich nanodroplets could form and act as a reservoir to maintain the maximum free drug concentration in solution, facilitating the absorption of the drug in vivo. Dissolution behavior of ASD has received increasing interests. This review will focus on the recent advances in ASD dissolution, including the generation and maintenance of supersaturated drug solution in absence or presence of liquid-liquid phase separation. Mechanism of drug release from ASD including polymer-controlled dissolution and drug-controlled dissolution will be introduced. Formation of amorphous drug-rich nanodroplets during dissolution and the underlying mechanism will be discussed. Phase separation morphology of hydrated ASD plays a critical role in dissolution behavior of ASD, which will be highlighted. Supersaturated drug solution shows poor physical stability and tends to crystallize. The effect of polymer and surfactant on supersaturated drug solution will be demonstrated and some unexpected results will be shown. Physicochemical properties of drug and polymer could impact ASD dissolution and some of them even show opposite effect on dissolution and physical stability of ASD in solid state, respectively. This review will contribute to a better understanding of ASD dissolution and facilitate a rational design of ASD formulation.
Collapse
|
40
|
Deac A, Qi Q, Indulkar AS, Purohit HS, Gao Y, Zhang GGZ, Taylor LS. Dissolution Mechanisms of Amorphous Solid Dispersions: Role of Drug Load and Molecular Interactions. Mol Pharm 2023; 20:722-737. [PMID: 36545917 DOI: 10.1021/acs.molpharmaceut.2c00892] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
High drug load amorphous solid dispersions (ASDs) have been a challenge to formulate partially because drug release is inhibited at high drug loads. The maximum drug load prior to inhibition of release has been termed the limit of congruency (LoC) and has been most widely studied for copovidone (PVPVA)-based ASDs. The terminology was derived from the observation that below LoC, the polymer controlled the kinetics and the drug and the polymer released congruently, while above LoC, the release rates diverged and were impaired. Recent studies show a correlation between the LoC value and drug-polymer interaction strength, where a lower LoC was observed for systems with stronger interactions. The aim of this study was to investigate the causality between drug-PVPVA interaction strength and LoC. Four chemical analogues with diverse abilities to interact with PVPVA were used as model drugs. The distribution of the polymer between the dilute aqueous phase and the insoluble nanoparticles containing drug was studied with solution nuclear magnetic resonance spectroscopy and traditional separation techniques to understand the thermodynamics of the systems in a dilute environment. Polymer diffusion to and from ASD particles suspended in aqueous solution was monitored for drug loads above the LoC to investigate the thermodynamic driving force for polymer release. The surface composition of ASD compacts before and after exposure to buffer was studied with Fourier transform infrared spectroscopy to capture potential kinetic barriers to release. It was found that ASD compacts with drug loads above the LoC formed an insoluble barrier on the surface that was in pseudo-equilibrium with the aqueous phase and prevented further release of drugs and polymers during dissolution. The insoluble barrier contained a substantial amount of the polymer for the strongly interacting drug-polymer systems. In contrast, a negligible amount was found for the weakly interacting systems. This observation provides an explanation for the ability of strongly interacting systems to form an insoluble barrier at lower drug loads. The study highlights the importance of thermodynamic and kinetic factors on the dissolution behavior of ASDs and provides a potential framework for maximizing the drug load in ASDs.
Collapse
Affiliation(s)
- Alexandru Deac
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana47907, United States
| | - Qingqing Qi
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana47907, United States
| | - Anura S Indulkar
- Development Sciences, Research and Development, AbbVie Inc, North Chicago, Illinois60064, United States
| | - Hitesh S Purohit
- Development Sciences, Research and Development, AbbVie Inc, North Chicago, Illinois60064, United States
| | - Yi Gao
- Development Sciences, Research and Development, AbbVie Inc, North Chicago, Illinois60064, United States
| | - Geoff G Z Zhang
- Development Sciences, Research and Development, AbbVie Inc, North Chicago, Illinois60064, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana47907, United States
| |
Collapse
|
41
|
Moseson DE, Benson EG, Cao Z, Bhalla S, Wang F, Wang M, Zheng K, Narwankar PK, Simpson GJ, Taylor LS. Impact of Aluminum Oxide Nanocoating on Drug Release from Amorphous Solid Dispersion Particles. Mol Pharm 2023; 20:593-605. [PMID: 36346665 DOI: 10.1021/acs.molpharmaceut.2c00818] [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: 11/09/2022]
Abstract
Atomic layer coating (ALC) is emerging as a particle engineering strategy to inhibit surface crystallization of amorphous solid dispersions (ASDs). In this study, we turn our attention to evaluating drug release behavior from ALC-coated ASDs, and begin to develop a mechanistic framework. Posaconazole/hydroxypropyl methylcellulose acetate succinate was used as a model system at both 25% and 50% drug loadings. ALC-coatings of aluminum oxide up to 40 nm were evaluated for water sorption kinetics and dissolution performance under a range of pH conditions. Scanning electron microscopy with energy dispersive X-ray analysis was used to investigate the microstructure of partially released ASD particles. Coating thickness and defect density (inferred from deposition rates) were found to impact water sorption kinetics. Despite reduced water sorption kinetics, the presence of a coating was not found to impact dissolution rates under conditions where rapid drug release was observed. Under slower releasing conditions, underlying matrix crystallization was reduced by the coating, enabling greater levels of drug release. These results demonstrate that water was able to penetrate through the ALC coating, hydrating the amorphous solid, which can initiate dissolution of drug and/or polymer (depending on pH conditions). Swelling of the ASD substrate subsequently occurs, disrupting and cracking the coating, which serves to facilitate rapid drug release. Water sorption kinetics are highlighted as a potential predictive tool to investigate the coating quality and its potential impact on dissolution performance. This study has implications for formulation design and evaluation of ALC-coated ASD particles.
Collapse
Affiliation(s)
- Dana E Moseson
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Emily G Benson
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ziyi Cao
- Department of Chemistry, College of Science, Purdue University, West Lafayette, Indiana 47907, United States
| | - Shradha Bhalla
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Fei Wang
- Applied Materials, Inc., 3100 Bowers Ave, Santa Clara, California 95054, United States
| | - Miaojun Wang
- Applied Materials, Inc., 3100 Bowers Ave, Santa Clara, California 95054, United States
| | - Kai Zheng
- Applied Materials, Inc., 3100 Bowers Ave, Santa Clara, California 95054, United States
| | - Pravin K Narwankar
- Applied Materials, Inc., 3100 Bowers Ave, Santa Clara, California 95054, United States
| | - Garth J Simpson
- Department of Chemistry, College of Science, Purdue University, West Lafayette, Indiana 47907, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
42
|
Moseson DE, Hiew TN, Su Y, Taylor LS. Formulation and Processing Strategies which Underpin Susceptibility to Matrix Crystallization in Amorphous Solid Dispersions. J Pharm Sci 2023; 112:108-122. [PMID: 35367246 DOI: 10.1016/j.xphs.2022.03.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 11/18/2022]
Abstract
Through matrix crystallization, an amorphous solid may transform directly into its more stable crystalline state, reducing the driving force for dissolution. Herein, the mechanism of matrix crystallization in an amorphous solid dispersion (ASD) was probed. ASDs of bicalutamide/copovidone were prepared by solvent evaporation and hot melt extrusion, and sized by mortar and pestle or cryomilling techniques, modulating the level of mechanical activation experienced by the sample. Drug loading (DL) of the binary ASD was varied from 5-50%, and ternary systems were formulated at 30% DL with two surfactants (sodium dodecyl sulfate, Vitamin E TPGS). Imaging of partially dissolved or crystallized compacts by scanning electron microscopy with energy-dispersive X-ray analysis and confocal fluorescence microscopy was performed to investigate pathways of hydration, phase separation, and crystallization. Monitoring drug and polymer release of ASD powder under non-sink conditions provided insight into supersaturation and desupersaturation profiles. Systems at the greatest risk of matrix crystallization had high DLs, underwent mechanical activation, and/or contained surfactant. Systems having greatest resistance to matrix crystallization had rapid and congruent drug and polymer release. This study has implications for formulation and process design of ASDs and risk assessment of matrix crystallization.
Collapse
Affiliation(s)
- Dana E Moseson
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Tze Ning Hiew
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Yongchao Su
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States; Analytical Research and Development, Merck & Co., Inc., Rahway, NJ 07065, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States.
| |
Collapse
|
43
|
Considerations in the Development of Physically Stable High Drug Load API- Polymer Amorphous Solid Dispersions in the Glassy State. J Pharm Sci 2023; 112:8-18. [PMID: 35948156 DOI: 10.1016/j.xphs.2022.08.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/04/2022] [Accepted: 08/04/2022] [Indexed: 02/05/2023]
Abstract
In this Commentary, the authors expand on their earlier studies of the solid-state long-term isothermal crystallization of amorphous API from the glassy state in amorphous solid dispersions, and focus on the effects of polymer concentration, and its implications for producing high load API doses with minimum polymer concentration. After presenting an overview of the various mechanistic factors which influence the ability of polymers to inhibit API crystallization, including the chemical structure of the polymer relative to the API, the nature and strength of API-polymer noncovalent interactions, polymer molecular weight, impact on primary diffusive molecular mobility, as well as on secondary motions in the bulk and surface phases of the glass, we consider in more detail, the effects of polymer concentration. Here, we examine the factors that appear to allow relatively low polymer concentrations, i.e., less than 10%w/w polymer, to greatly reduce crystallization, including a focus on the heterogeneous structure of the glassy state, and the possible spatial distribution and concentration of polymer in certain key regions of the glass. This is followed by a review and analysis of examples in the recent literature focused on determining the minimum polymer concentration in an amorphous solid dispersion, capable of producing optimally stable high drug load amorphous dispersions.
Collapse
|
44
|
Zhao P, Han W, Shu Y, Li M, Sun Y, Sui X, Liu B, Tian B, Liu Y, Fu Q. Liquid-liquid phase separation drug aggregate: Merit for oral delivery of amorphous solid dispersions. J Control Release 2023; 353:42-50. [PMID: 36414193 DOI: 10.1016/j.jconrel.2022.11.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022]
Abstract
As a promising strategy, amorphous solid dispersion has been extensively employed in improving the oral bioavailability of insoluble drugs. Despite the numerous advantages, the problems associated with supersaturation stability limit its further application. Recently, the formation and stability of the liquid-liquid phase separation drug aggregate (LLPS-DA) have been found to be vital for supersaturation maintenance. An in-depth review of LLPS-DA was required to further explore the supersaturation maintenance mechanism in vivo. Hence, this study aimed to present a short review to introduce the LLPS-DA, highlight the in vivo advantages for oral administration, and discuss the prospects to help understand the in vivo behavior of LLPS-DA.
Collapse
Affiliation(s)
- Peixu Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Wen Han
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Yecheng Shu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Mo Li
- Liaoning Institute for Drug Control, No. 7 Chongshan West Road, Shenyang 110016, China
| | - Yichi Sun
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Xiaofan Sui
- Liaoning Institute for Drug Control, No. 7 Chongshan West Road, Shenyang 110016, China
| | - Bingyang Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Baocheng Tian
- School of Pharmacy, Binzhou Medical University, No. 346, Guanhai Road, Yantai 264003, China
| | - Yanhua Liu
- Department of Pharmaceutics, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
| | - Qiang Fu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
45
|
Combining drug salt formation with amorphous solid dispersions - a double edged sword. J Control Release 2022; 352:47-60. [PMID: 36206947 PMCID: PMC9733678 DOI: 10.1016/j.jconrel.2022.09.056] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 09/26/2022] [Indexed: 11/08/2022]
Abstract
Glass transition temperature (Tg) is important for amorphous compounds because it can have implications on their physical and chemical stability. With drugs that possess ionizable acidic or basic groups, salt formation is a potential strategy to reduce re-crystallization tendency through Tg elevation. While salt formation has been reported to impact re-crystallization tendency, it is not known if this holds true for all drugs and if it is useful in the context of amorphous solid dispersion (ASD) formulations. In addition, little information on the impact of salt formation on drug release performance of ASD is available. Herein, the influence of salt formation and Tg elevation on the release performance of lumefantrine (Tg = 19.7 °C) when formulated as an ASD with copovidone (PVPVA) was examined. Lumefantrine salts and lumefantrine salt-PVPVA ASDs with drug loadings (DLs) ranging from 5 to 30% were prepared. The acids used for salt formation were benzoic acid, benzenesulfonic acid, camphorsulfonic acid, hydrochloric acid, p-toluenesulfonic acid, poly(ethylene) glycol 250 diacid (PEG 250 diacid), and sulfuric acid. Salt formation resulted in an elevation of Tg compared to lumefantrine free base, with the largest increase in Tg observed with lumefantrine sulfate. With a lower Tg salt, ASDs could be formulated at higher DLs while ensuring drug release. In contrast, drug release ceased at a DL as low as 5% when Tg of the salt was high. However, ASDs with lower Tgs such as the benzoate and PEG 250 diacid salts showed poor stability against re-crystallization when stored under stress storage conditions. When using a salt in an ASD formulation, attention should be paid to the Tg of the salt, since it may show opposing effects on physical stability and drug release, at least for PVPVA-based ASDs.
Collapse
|
46
|
Tailored Supersaturable Immediate Release Behaviors of Hypotensive Supersaturating Drug-Delivery Systems Combined with Hot-Melt Extrusion Technique and Self-Micellizing Polymer. Polymers (Basel) 2022; 14:polym14224800. [PMID: 36432925 PMCID: PMC9693352 DOI: 10.3390/polym14224800] [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: 09/22/2022] [Revised: 10/31/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
The short-term immediate release of supersaturated drug-delivery systems (SDDSs) presents an interesting process that can be tailored to multi-stage release events including initial release after dosing and dissolution, evolved release over longer dissolution periods for biological absorption, and terminal release following the end of immediate release. However, although comprehensive analysis of these critical release behaviors is often ignored yet essential for understanding the supersaturable immediate-release events for supersaturable solid formations when employing new techniques or polymers matched to a particular API. Hot-melt extrusion (HME) has become a popular continuous thermodynamic disordering technique for amorphization. The self-micellizing polymer Soluplus® is reported to be a potential amorphous and amphiphilic graft copolymer frequently used in many nano/micro supersaturable formulations. Our current work aims to develop hypotensive supersaturating solid dispersion systems (faSDDSHME) containing the BCS II drug, felodipine, when coordinately employing the HME technique and self-micellizing Soluplus®, and to characterize their amorphization as well as immediate release. Other discontinuous techniques were used to prepare control groups (faSDDSSE and faSDDSQC). Tailored initial/evolved/terminal three-stage supersaturable immediate-release behaviors were identified and possible mechanisms controlling the release were explored. HME produced the highest initial release in related faSDDSHME. During the evolved-release period, highly extended "spring-parachute" process was found in HME-induced amorphization owing to its superior supersaturation duration. Due to the enhanced crystallization inhibition effect, faSDDSHME displayed the strongest terminal release as measured by solubility. For release mechanisms associated with HME, molecular interaction is not the likely dominant mechanism responsible for the improved properties induced by faSDDSHME. For release mechanisms involved with the polymer Soluplus® itself, they were found to inhibit drug recrystallization, spontaneously solubilize the drug and lead to improved molecular interactions in all SDDS systems, which were the factors responsible for the improved release. These mechanisms play an important role for the generation of an extended multi-stage immediate release produced via HME or self-micellizing polymer. This study provides a deeper understanding on amorphization and superior multi-stage supersaturable immediate-release behaviors for a particular hypotensive supersaturated delivery system combined with an HME-based continuous manufacturing technique and self-micellizing polymer strategy.
Collapse
|
47
|
Moseson DE, Benson EG, Nguyen HT, Wang F, Wang M, Zheng K, Narwankar PK, Taylor LS. Atomic Layer Coating to Inhibit Surface Crystallization of Amorphous Pharmaceutical Powders. ACS APPLIED MATERIALS & INTERFACES 2022; 14:40698-40710. [PMID: 36054111 DOI: 10.1021/acsami.2c12666] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Preventing crystallization is a primary concern when developing amorphous drug formulations. Recently, atomic layer coatings (ALCs) of aluminum oxide demonstrated crystallization inhibition of high drug loading amorphous solid dispersions (ASDs) for over 2 years. The goal of the current study was to probe the breadth and mechanisms of this exciting finding through multiple drug/polymer model systems, as well as particle and coating attributes. The model ASD systems selected provide for a range of hygroscopicity and chemical functional groups, which may contribute to the crystallization inhibition effect of the ALC coatings. Atomic layer coating was performed to apply a 5-25 nm layer of aluminum oxide or zinc oxide onto ASD particles, which imparted enhanced micromeritic properties, namely, reduced agglomeration and improved powder flowability. ASD particles were stored at 40 °C and a selected relative humidity level between 31 and 75%. Crystallization was monitored by X-ray powder diffraction and scanning electron microscopy (SEM) up to 48 weeks. Crystallization was observable by SEM within 1-2 weeks for all uncoated samples. After ALC, crystallization was effectively delayed or completely inhibited in some systems up to 48 weeks. The delay achieved was demonstrated regardless of polymer hygroscopicity, presence or absence of hydroxyl functional groups in drugs and/or polymers, particle size, or coating properties. The crystallization inhibition effect is attributed primarily to decreased surface molecular mobility. ALC has the potential to be a scalable strategy to enhance the physical stability of ASD systems to enable high drug loading and enhanced robustness to temperature or relative humidity excursions.
Collapse
Affiliation(s)
- Dana E Moseson
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Emily G Benson
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Hanh Thuy Nguyen
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Fei Wang
- Applied Materials, Inc., 3100 Bowers Avenue, Santa Clara, California 95054, United States
| | - Miaojun Wang
- Applied Materials, Inc., 3100 Bowers Avenue, Santa Clara, California 95054, United States
| | - Kai Zheng
- Applied Materials, Inc., 3100 Bowers Avenue, Santa Clara, California 95054, United States
| | - Pravin K Narwankar
- Applied Materials, Inc., 3100 Bowers Avenue, Santa Clara, California 95054, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
48
|
Shi Q, Chen H, Wang Y, Wang R, Xu J, Zhang C. Amorphous Solid Dispersions: Role of the Polymer and Its Importance in Physical Stability and In Vitro Performance. Pharmaceutics 2022; 14:pharmaceutics14081747. [PMID: 36015373 PMCID: PMC9413000 DOI: 10.3390/pharmaceutics14081747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/15/2022] [Accepted: 08/18/2022] [Indexed: 01/25/2023] Open
Abstract
Amorphous solid dispersions stabilized by one or more polymer(s) have been widely used for delivering amorphous drugs with poor water solubilities, and they have gained great market success. Polymer selection is important for preparing robust amorphous solid dispersions, and considerations should be given as to how the critical attributes of a polymer can enhance the physical stability, and the in vitro and in vivo performances of a drug. This article provides a comprehensive overview for recent developments in the understanding the role of polymers in amorphous solid dispersions from the aspects of nucleation, crystal growth, overall crystallization, miscibility, phase separation, dissolution, and supersaturation. The critical properties of polymers affecting the physical stability and the in vitro performance of amorphous solid dispersions are also highlighted. Moreover, a perspective regarding the current research gaps and novel research directions for better understanding the role of the polymer is provided. This review will provide guidance for the rational design of polymer-based amorphous pharmaceutical solids with desired physicochemical properties from the perspective of physical stability and in vitro performance.
Collapse
Affiliation(s)
- Qin Shi
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng 224005, China
- Correspondence: (Q.S.); (C.Z.)
| | - Haibiao Chen
- Institute of Marine Biomedicine, Shenzhen Polytechnic, Shenzhen 518055, China
| | - Yanan Wang
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Ruoxun Wang
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Jia Xu
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Chen Zhang
- Institute of Marine Biomedicine, Shenzhen Polytechnic, Shenzhen 518055, China
- Correspondence: (Q.S.); (C.Z.)
| |
Collapse
|
49
|
Role of surfactants in improving release from higher drug loading amorphous solid dispersions. Int J Pharm 2022; 625:122120. [PMID: 35987321 DOI: 10.1016/j.ijpharm.2022.122120] [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: 05/04/2022] [Revised: 08/08/2022] [Accepted: 08/12/2022] [Indexed: 11/21/2022]
Abstract
Amorphous solid dispersion formulations (ASD) are increasingly being used as a formulation strategy to improve bioavailability of poorly soluble drugs. One of the limitations of ASDs, in particular for high glass transition temperature (Tg) compounds, is the drug loading threshold (termed the limit of congruency, LoC) below which rapid, complete and congruent release of drug and polymer is achieved. In this study, several ionic and non-ionic surfactants were added to atazanavir-copovidone ASDs with the main goal of increasing the limit of congruency. Atazanavir (ATZ) is a relatively high Tg compound with a LoC of 5 % drug loading (DL). Surface normalized dissolution studies revealed that addition of 5 % w/w of surfactant, sodium dodecyl sulfate (SDS) or cetrimonium bromide (CTAB), to the binary copovidone-based ASD doubled the LoC (from 5 to 10 % DL), resulting in a more than 30-fold increase in total release compared to the corresponding binary ASD. Moreover, addition of 5 % of Span®80 increased the LoC to 15 % DL. ASD Tg was found to decrease upon addition of surfactants and water sorption extent was found to increase. We speculate that surfactants act as plasticizers, which may facilitate polymer release from ASDs containing a high Tg drug, providing a possible explanation for the observed enhancement in drug release from ternary ASDs and the increase in LoC.
Collapse
|
50
|
Qi Q, Taylor LS. Improved dissolution of an enteric polymer and its amorphous solid dispersions by polymer salt formation. Int J Pharm 2022; 622:121886. [PMID: 35661745 DOI: 10.1016/j.ijpharm.2022.121886] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 11/29/2022]
Abstract
Weakly acidic polymers, historically used as enteric coatings, are increasingly being employed in solubility-enhancing amorphous solid dispersion (ASD) formulations. However, there is a lack of fundamental understanding around how these carboxylic acid-containing polymers dissolve, in particular when molecularly mixed with a lipophilic drug, as in an ASD. Identification of critical factors dominating their dissolution is vital for rational design of new polymers with enhanced release properties to address contemporary ASD delivery challenges, notably achieving good release at higher drug loadings. Herein, after identification of polymer solubilization via ionization as the rate limiting step for dissolution, hydroxypropylmethyl cellulose phthalate (HP-50) was converted to a salt by neutralization of the phthalic acid groups with different bases. Surface normalized dissolution was performed to assess the dissolution rate improvement achieved by polymer pre-ionization via salt formation. Polymer salts showed ∼ 3-fold faster release than HP-50 at pH 6.8 (50 mM sodium phosphate buffer). Importantly, a polymer salt was able to maintain a rapid dissolution rate, irrespective of the buffer capacity of the medium, whereas the protonated polymer showed greatly diminished dissolution as medium buffer capacity decreased toward physiological gastrointestinal tract values. HP-50 and two polymer salts were formulated into ASDs with miconazole, a lipophilic and weakly basic antifungal drug, at a 20% drug loading. Rapid drug release rates were achieved with polymer salt ASDs, whereby drug release was 14 times faster than from the protonated HP-50 ASD. This study highlights the critical role of polymer ionization and buffer capacity in the dissolution of HP-50-based systems and how pre-ionization via polymer salt formation is a successful strategy for the design of new polymers for improved ASD performance.
Collapse
Affiliation(s)
- Qingqing Qi
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| |
Collapse
|