1
|
Luo M, Chen A, Shan S, Guo M, Cai T. Molar Ratio-Dependent Crystallization in Coamorphous Celecoxib-Carbamazepine Systems: The Interplay of Thermodynamics and Kinetics. Mol Pharm 2025. [PMID: 40360458 DOI: 10.1021/acs.molpharmaceut.5c00278] [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: 05/15/2025]
Abstract
Coamorphous drug delivery systems have emerged as a promising formulation strategy to enhance the solubility, oral bioavailability, and physical stability of poorly water-soluble drugs. The molar ratio of components in coamorphous systems plays a critical role in determining their physical stability. In this study, we investigated the crystallization behavior of coamorphous celecoxib-carbamazepine (CEL-CBZ) systems at different molar ratios. The growth rates of CEL crystals, CBZ crystals, and CEL-CBZ cocrystals were observed to exhibit distinct dependencies on the molar ratio of coamorphous systems, primarily due to their unique thermodynamic driving forces, despite sharing the same kinetic factor. The influence of the molar ratio on the crystallization of coamorphous systems arises from the interplay between its effects on molecular mobility and thermodynamic driving forces, leading to either cooperative or competing effects. Both the crystal growth and crystallization tendency results reveal that thermodynamics plays a more dominant role than kinetics in the crystallization of coamorphous CEL-CBZ systems across various molar ratios. This study provides fundamental insights into the mechanism by which the molar ratio influences the crystallization of coamorphous systems, highlighting the complex crystallization behavior of multicomponent amorphous systems as an interplay between kinetics and thermodynamics.
Collapse
Affiliation(s)
- Minqian Luo
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - An Chen
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Shiyu Shan
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Minshan Guo
- 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
|
2
|
Ramachandran G, Chacko IA, Mishara MG, Khopade AJ, Sabitha M, Sudheesh MS. A review on design rules for formulating amorphous solid dispersions based on drug-polymer interactions in aqueous environment. Int J Pharm 2025; 675:125541. [PMID: 40164414 DOI: 10.1016/j.ijpharm.2025.125541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/09/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Amorphous solid dispersions (ASDs) are multi-component formulations in which a drug is molecularly dispersed in a carrier. ASDs undergo complex dissolution mechanisms to generate and sustain a supersaturated state of poorly soluble drugs. The link between enhanced solubility, supersaturation stability and drug-polymer interaction (DPI) is critical for the rational design of ASDs. The key mechanism responsible for a high bioavailability is the evolution of supersaturation during the dissolution of ASDs which is also the driving force for drug precipitation. A critical determinant of robust supersaturation generation and stability during dissolution is the molecular interaction between the drug and polymer. Characterization of DPI in a solution state is, however, challenging because of the poor hydrodynamic resolution of the techniques, traditionally used in solid-state analysis. Further, the dissolution conditions, such as the choice of buffer, pH and ionic strength may complicate the analyses and predictions. The role of DPI is a poorly understood aspect of ASD dissolution and therefore is an active area of research. DPI is critical for understanding the design rules for formulating an optimal ASD formulation. The review focuses on different aspects of DPI to stabilize the supersaturated state of a drug during the dissolution of ASDs.
Collapse
Affiliation(s)
- Gayathri Ramachandran
- Molecular Pharmaceutics and Biopharmaceutics Research Lab (MPBRL), Dept. of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi 682041, India
| | - Indhu Annie Chacko
- Molecular Pharmaceutics and Biopharmaceutics Research Lab (MPBRL), Dept. of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi 682041, India
| | - M G Mishara
- Molecular Pharmaceutics and Biopharmaceutics Research Lab (MPBRL), Dept. of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi 682041, India
| | - Ajay Jaysingh Khopade
- Department of Formulation R&D Non-Orals, Sun Pharmaceutical Industries Ltd., Vadodara, India
| | - M Sabitha
- Molecular Pharmaceutics and Biopharmaceutics Research Lab (MPBRL), Dept. of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi 682041, India
| | - M S Sudheesh
- Molecular Pharmaceutics and Biopharmaceutics Research Lab (MPBRL), Dept. of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi 682041, India.
| |
Collapse
|
3
|
Cools L, Van den Mooter G. A comprehensive overview of the role of intermolecular interactions in amorphous solid dispersions. Int J Pharm 2025; 674:125441. [PMID: 40089043 DOI: 10.1016/j.ijpharm.2025.125441] [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: 01/11/2025] [Revised: 02/25/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
Many recent studies have indicated that drug-polymer intermolecular interactions are an important aspect of amorphous solid dispersions (ASDs) and determine many of the properties of this type of formulations. In this review, a comprehensive overview is given of the latest insights with respect to intermolecular interactions in ASDs. The thermodynamic properties and theoretical considerations of the interactions are discussed, followed by a detailed and critical overview of the various solid-state analysis techniques used to probe interactions at the disposal of the formulation scientist. As the physical stability and the pharmaceutical performance of the ASD are its most crucial properties, the most recent understanding of the influence of drug-polymer interactions on these aspects is addressed as well. It is clear that intermolecular interactions may provide many advantages for ASDs but need to be weighed against the possible disadvantages. Further investigation into the interplay and trade-off between physical stability and dissolution properties is necessary in order to be able to take full advantage of the possible benefits of the interactions.
Collapse
Affiliation(s)
- Lennert Cools
- Drug Delivery and Disposition, KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Campus Gasthuisberg ON2, Herestraat 49 b921 3000 Leuven, Belgium; Applied and Analytical Chemistry, NMR Group, Institute for Materials Research (imo-imomec), UHasselt, 3590 Diepenbeek, Belgium
| | - Guy Van den Mooter
- Drug Delivery and Disposition, KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Campus Gasthuisberg ON2, Herestraat 49 b921 3000 Leuven, Belgium.
| |
Collapse
|
4
|
Lange J, Senniksen MB, Wyttenbach N, Page S, Bateman LM, O’Dwyer PJ, Saal W, Kuentz M, Griffin BT. Mechanistic Investigation into the Phase Separation Behavior of Soluplus in the Presence of Biorelevant Media. Mol Pharm 2025; 22:1958-1972. [PMID: 40066684 PMCID: PMC11979885 DOI: 10.1021/acs.molpharmaceut.4c01140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 02/13/2025] [Accepted: 02/13/2025] [Indexed: 04/08/2025]
Abstract
More than a decade since its introduction, the polymeric excipient Soluplus continues to receive considerable attention for its application in the development of amorphous solid dispersions (ASDs) and its utility as a solubilizer for drugs exhibiting solubility limited absorption. While it is well-recognized that Soluplus forms micelles, the impact of its lower critical solution temperature of approximately 40 °C remains an underexplored aspect. This study investigated the phase behavior of Soluplus in fasted-state simulated intestinal fluid (FaSSIF-V1). It was demonstrated that Soluplus forms a dispersed polymer-rich coacervate phase, which coexists with Soluplus micelles at 37 °C. This behavior was confirmed by cloud point measurements, visually discernible phases after centrifugation, as well as multi-angle dynamic light scattering (MADLS) measurements, and quantitative 1H-nuclear magnetic resonance (NMR) spectroscopy of Soluplus concentrations in the supernatant pre- and post-centrifugation. The practical relevance of these findings was contextualized by solvent shift experiments and dissolution testing of spray-dried ASD. The results demonstrated that the poorly water-soluble drug RO6897779 resided in a polymer-rich coacervate phase and was spun down during centrifugation, which resulted in an amorphous pellet exhibiting the characteristics of a viscous liquid. The entrapment of the drug within the polymer-rich phase was further analyzed by temperature- and time-dependent MADLS experiments. The findings of this study are of particular relevance for a mechanistic understanding, relevant to comprehending in vitro-in vivo relationships of Soluplus-based ASDs. Low sampled drug concentrations in FaSSIF-V1 at 37 °C may originate not only from limited drug release and precipitation but also from the formation of a drug-containing, polymer-rich Soluplus phase. Therefore, a liquid-liquid phase separation occurring from Soluplus-based formulations in a biorelevant medium can be excipient-driven, which is different from the common perception that phase separation in the solution state is triggered primarily by high drug concentrations exceeding their amorphous solubility.
Collapse
Affiliation(s)
- Justus
Johann Lange
- School
of Pharmacy, University College Cork, College Road, Cork County, T12 R229 Cork , Ireland
| | - Malte Bøgh Senniksen
- Pharmaceutical
R&D, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
- Fraunhofer
Institute for Translational Medicine and Pharmacology, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Nicole Wyttenbach
- Roche
Pharma Research and Early Development, Therapeutic Modalities, Roche
Innovation Center Basel, F. Hoffmann-La
Roche Ltd., Grenzacherstrasse
124, 4070 Basel, Switzerland
| | - Susanne Page
- Pharmaceutical
R&D, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Lorraine M. Bateman
- School
of Pharmacy, University College Cork, College Road, Cork County, T12 R229 Cork , Ireland
- Analytical
& Biological Research Facility, University
College Cork, College
Road, T12 YN60 Cork, Ireland
| | - Patrick J. O’Dwyer
- School
of Pharmacy, University College Cork, College Road, Cork County, T12 R229 Cork , Ireland
| | - Wiebke Saal
- Roche
Pharma Research and Early Development, Therapeutic Modalities, Roche
Innovation Center Basel, F. Hoffmann-La
Roche Ltd., Grenzacherstrasse
124, 4070 Basel, Switzerland
| | - Martin Kuentz
- Institute
of Pharma Technology, University of Applied
Sciences and Arts Northwestern Switzerland, Hofackerstrasse 30, CH-4132 Muttenz, Switzerland
| | - Brendan T. Griffin
- School
of Pharmacy, University College Cork, College Road, Cork County, T12 R229 Cork , Ireland
| |
Collapse
|
5
|
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
|
6
|
Fan F, Zhou F, Zhang J, Yang J, Zhuang K, Shan Y, Jiang L, Zhang J. Developing Soluplus®-Based Microparticle Amorphous Solid Dispersions with High Drug Loading for Enhanced Celecoxib Dissolution via Electrospraying. AAPS PharmSciTech 2025; 26:47. [PMID: 39881034 DOI: 10.1208/s12249-025-03041-7] [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] [Accepted: 01/08/2025] [Indexed: 01/31/2025] Open
Abstract
Amorphous solid dispersion (ASD) is one of the most studied strategies for improving the dissolution performance of poorly water-soluble drugs, but ASDs often have low drug loadings, thereby necessitating larger dosage sizes. This study intended to create Soluplus® (SOL)-based microparticle ASDs with high drug loading (up to 60 w/w%) and long-term stability (at least 16 months) using electrospraying to enhance the dissolution of poorly water-soluble celecoxib (CEL). X-ray diffraction (XRD) and differential scanning calorimetry (DSC) analyses showed that the electrosprayed SOL-CEL microparticles were amorphous, and Fourier transform infrared spectroscopy (FTIR) data indicated the presence of hydrogen bonding between SOL and CEL in the microparticles, which helped stabilize the ASDs. In vitro dissolution studies demonstrated that these ASDs improved the CEL dissolution rate by up to 8.2-fold compared to the crystalline form. Electrospraying presents a promising alternative to conventional methods like hot-melt extrusion (HME) and spraying drying (SD) for the production of ASDs, providing simplicity, high drug loading capability and long-term stability, thus catering to a variety of poorly water-soluble drugs.
Collapse
Affiliation(s)
- Fan Fan
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| | - Feng Zhou
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
| | - Jiayu Zhang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Junhui Yang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, China
| | - Kai Zhuang
- Pharma Solutions, Nutrition and Health, BASF (China) Company, Ltd., 333 Jiang Xin Sha Road, Shanghai, 200137, China
| | - Yudong Shan
- Hangzhou Zhongmeihuadong Pharmaceutical Co., Ltd., 866 Moganshan Road, Hangzhou, 310011, China
| | - Lei Jiang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China.
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China.
| | - Jiantao Zhang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, China.
- Ningbo Cixi Institute of Biomedical Engineering, Cixi, 315300, China.
| |
Collapse
|
7
|
Garbiec E, Rosiak N, Sip S, Zalewski P, Cielecka-Piontek J. Curcumin Solubility and Bioactivity Enhancement Through Amorphization with Tryptophan via Supercritical Fluid Technology. Int J Mol Sci 2025; 26:855. [PMID: 39859569 PMCID: PMC11766122 DOI: 10.3390/ijms26020855] [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: 11/15/2024] [Revised: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Curcumin, a compound known for its antioxidant and neuroprotective properties, faces challenges due to its low water solubility, which can limit its effectiveness. One effective method to address this issue is through amorphization. Incorporating curcumin into a polymeric matrix to form amorphous solid dispersions is a common approach. Another strategy involves co-amorphous systems, where low-molecular-weight components act as co-formers. A recent innovative approach combines these strategies. This study used tryptophan as a co-former and prepared systems using supercritical fluid technology. The amorphous nature of two systems was confirmed through X-ray powder diffraction: one with 10% curcumin and a polymer, and another with 10% curcumin, a polymer, and tryptophan. Fourier-transform infrared analysis demonstrated molecular interactions among all components in the systems. Scanning electron microscopy revealed that the amorphization process significantly modified the morphology of the powder particles. The ternary system with tryptophan notably increased curcumin solubility by over 300-fold. The amorphous form of curcumin in both systems exhibited significantly higher dissolution rates compared to its crystalline form. The system with tryptophan showed more than a threefold improvement in permeability according to the PAMPA test. The enhanced solubility led to over a sixfold increase in antioxidant activity and a 25-fold improvement in the inhibition of the enzyme butyrylcholinesterase.
Collapse
Affiliation(s)
| | | | | | | | - Judyta Cielecka-Piontek
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3 Str., 60-806 Poznan, Poland; (E.G.); (N.R.); (S.S.); (P.Z.)
| |
Collapse
|
8
|
Bapat P, Taylor LS. Impact of HPMCAS Grade on the Release of Weakly Basic Drugs from Amorphous Solid Dispersions. Mol Pharm 2025; 22:397-407. [PMID: 39704640 DOI: 10.1021/acs.molpharmaceut.4c00986] [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] [Indexed: 12/21/2024]
Abstract
Oppositely charged species can form electrostatic interactions in aqueous solution, and these may lead to reduced solubility of the interacting components. Herein, insoluble complex formation between the lipophilic weakly basic drugs, cinnarizine or loratadine, and the enteric polymer, hydroxypropyl methylcellulose acetate succinate (HPMCAS), was studied and used to better understand drug and polymer release from their corresponding amorphous solid dispersions (ASDs). Surface area normalized release experiments were performed at various pH conditions for three different grades of HPMCAS, LF, MF and HF, as well as their ASDs. Both polymer and drug release rates were measured for the ASDs. Complexation tendency was evaluated by measuring the extent of polymer loss from the aqueous phase in the presence of the drug. Results showed that release from ASDs with HPMCAS-LF was less impacted by the presence of a cationic form of the drug than ASDs prepared with the HF grade. Furthermore, an increase in pH, leading to a reduction in the extent of ionized drug also led to an improvement in release rate. These observations provide a baseline to understand the role of drug-polymer electrostatic interactions on release from ASDs formulated with HPMCAS. Future studies should focus on adding complexity to media conditions by employing simulated intestinal fluids with solubilizing components.
Collapse
Affiliation(s)
- Pradnya Bapat
- 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
|
9
|
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
|
10
|
Borrmann D, Friedrich P, Smuda J, Sadowski G. Counteracting the loss of release for indomethacin-copovidone ASDs. J Pharm Sci 2025; 114:449-457. [PMID: 39510502 DOI: 10.1016/j.xphs.2024.10.022] [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] [Revised: 10/13/2024] [Accepted: 10/13/2024] [Indexed: 11/15/2024]
Abstract
This work revisits the changing release behavior of indomethacin(IND)-copovidone amorphous solid dispersions (ASDs) when increasing their drug load (DL). While showing congruent release behavior at DL 0.1, ASDs with DLs of 0.3 and higher show incongruent release finally resulting in a complete loss of release. To study and explain this phenomenon, we modeled the release kinetics of these ASDs and looked into their phase behavior both experimentally and theoretically. We applied a diffusion model to accurately describe experimental release profiles for congruent release, incongruent release as well as for loss of release. Predicted concentration profiles for IND, copovidone, and water within the ASD revealed the formation of an ASD layer that almost exclusively contains amorphous IND. Our phase-diagram predictions and experimental data explain this phenomenon by water-induced phase separation in those parts of the ASD which did absorb water from the dissolution medium. Whereas the evolving copovidone-rich phase dissolved, the IND-rich phase remained undissolved and formed a super-hydrophobic cover of the remaining inner core of the ASD, thus finally completely preventing its dissolution. Higher DLs promote phase separation. This leads to the counterintuitive effect that the higher the DL, the lower the absolute amount of IND released. While the ASD containing 6 mg IND (DL 0.1) released 6 mg IND, the one containing 42 mg IND (DL 0.7) released only 1 mg IND. The theoretical approach applied in this work is for the first time able to quantitatively predict that reducing DL or tablet size could be used to overcome this problem.
Collapse
Affiliation(s)
- Dominik Borrmann
- Department of Chemical and Biochemical Engineering, Laboratory of Thermodynamics, TU Dortmund University, Figge-Str. 70, D-44227 Dortmund, Germany
| | - Pascal Friedrich
- Department of Chemical and Biochemical Engineering, Laboratory of Thermodynamics, TU Dortmund University, Figge-Str. 70, D-44227 Dortmund, Germany
| | - Justin Smuda
- Department of Chemical and Biochemical Engineering, Laboratory of Thermodynamics, TU Dortmund University, Figge-Str. 70, D-44227 Dortmund, Germany
| | - Gabriele Sadowski
- Department of Chemical and Biochemical Engineering, Laboratory of Thermodynamics, TU Dortmund University, Figge-Str. 70, D-44227 Dortmund, Germany.
| |
Collapse
|
11
|
Benson EG, Moseson DE, Bhalla S, Wang F, Wang M, Zheng K, Narwankar PK, Taylor LS. Dissolution of copovidone-based amorphous solid dispersions: Influence of atomic layer coating, hydration kinetics, and formulation. J Pharm Sci 2025; 114:323-335. [PMID: 39389537 DOI: 10.1016/j.xphs.2024.10.001] [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: 07/23/2024] [Revised: 10/02/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
Atomic layer coating (ALC) is an emerging, solvent-free technique to coat amorphous solid dispersion (ASD) particles with a nanolayer ceramic coating that has been shown to improve powder characteristics and limit drug crystallization. Herein, we evaluate the impact of aluminum oxide coatings with varying thickness and conformality on the release behavior of ritonavir/copovidone ASDs. Release performance of powders, neat tablets, and formulated tablets was studied. Confocal fluorescence microscopy (CFM) was used to visualize particle hydration and phase separation during immersion of the ASD in aqueous media. CFM revealed particle hydration requires defects for solvent penetration, but coatings, regardless of thickness, had minor impacts on powder dissolution provided defects were present. In tablets where less surface area is exposed to the dissolution media due to gel formation, slowed hydration kinetics resulted in phase separation of the drug from the polymer in coated samples, limiting release. Formulation with two superdisintegrants, crospovidone and croscarmellose sodium, as well as lactose achieved ∼90% release in less than 10 minutes, matching the uncoated ASD particles of the same formulation. This study highlights the importance of hydration rate, as well as the utility of confocal fluorescence microscopy to provide insight into release and phase behavior of ASDs.
Collapse
Affiliation(s)
- Emily G Benson
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Dana E Moseson
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Shradha Bhalla
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Fei Wang
- Applied Materials, Inc., 3100 Bowers Ave, Santa Clara, CA 95054, United States
| | - Miaojun Wang
- Applied Materials, Inc., 3100 Bowers Ave, Santa Clara, CA 95054, United States
| | - Kai Zheng
- Applied Materials, Inc., 3100 Bowers Ave, Santa Clara, CA 95054, United States
| | - Pravin K Narwankar
- Applied Materials, Inc., 3100 Bowers Ave, Santa Clara, CA 95054, United States
| | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
12
|
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
|
13
|
Yu M, Zhou D, Oberoi HS, Salem AH, McKee LA, Arnholt JR, Purohit HS, Law D. Scale-up and clinical bioavailability assessment of a 45% drug loaded amorphous nanoparticle formulation of a BCS IV compound for oral delivery. J Pharm Sci 2025; 114:383-393. [PMID: 39427713 DOI: 10.1016/j.xphs.2024.10.014] [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: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024]
Abstract
A 45 % drug loaded (DL) amorphous nanoparticle (ANP) formulation for a BCS IV drug demonstrated promising pharmacokinetics in dogs (Purohit, et al., J. Pharm. Sci. 2023(113)1007-1019). This preclinical data enabled a human proof-of-concept assessment opportunity. The ANP freeze dried powder for oral suspension was prepared using solvent/antisolvent precipitation followed by organic solvent removal and freeze drying (FD). Challenges manifested during scale-up from 50 g to 280 g. Given the preclinical data, formulation change was restricted, therefore, process modifications were implemented. Cold collection after precipitation prevented particle growth but resulted in 75 nm particles at clinical scale (CS), compared to 150 nm at laboratory scale (LS). This size decrease rendered stabilizer amounts suboptimal for FD operation. Consequently, when FD powder was resuspended in water a smaller fraction of particles was below 450 nm (by filtration), ∼65 % for CS compared to ∼85 % for LS. Formulation was stable for > 6 months, evaluated by monitoring moisture content, assay, powder X-ray diffraction (PXRD), and redispersion time. Despite ∼65 % re-dispersibility, this 45 % DL formulation in humans had higher Cmax and AUC ∼73 % and ∼46 % respectively in fasted-state, and under fed-state it met bioequivalence criteria for AUC but Cmax was 20 % lower compared to reference (10 % DL ASD tablets) demonstrating advantage of ANP strategy over ASD approach.
Collapse
Affiliation(s)
- Mengqi Yu
- Small molecule CMC development, Drug Product Development, AbbVie Inc., North Chicago, IL 60064, United States
| | - Deliang Zhou
- Small molecule CMC development, Drug Product Development, AbbVie Inc., North Chicago, IL 60064, United States
| | - Hardeep S Oberoi
- Small molecule CMC development, Drug Product Development, AbbVie Inc., North Chicago, IL 60064, United States
| | - Ahmed Hamed Salem
- Clinical Pharmacy, Ain Shams University, Cairo, Egypt; Clinical Pharmacology, AbbVie Inc., North Chicago, IL, USA
| | - Laura A McKee
- Process Research and development, Technical Operations, AbbVie Inc., North Chicago, IL, USA
| | - Jason R Arnholt
- Small molecule CMC development, Analytical Research and Development, AbbVie Inc., North Chicago, IL, USA
| | - Hitesh S Purohit
- Small molecule CMC development, Drug Product Development, AbbVie Inc., North Chicago, IL 60064, United States.
| | - Devalina Law
- Small molecule CMC development, Drug Product Development, AbbVie Inc., North Chicago, IL 60064, United States.
| |
Collapse
|
14
|
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
|
15
|
Zhang W, Thool P, Weitz BW, Hou HH. Investigating the effects of formulation variables on the disintegration of spray dried amorphous solid dispersion tablets. J Pharm Sci 2025; 114:304-312. [PMID: 39374694 DOI: 10.1016/j.xphs.2024.09.024] [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: 07/18/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/09/2024]
Abstract
Amorphous solid dispersion (ASD) tablets based on hydrophilic polymer carriers may encounter disintegration challenges. In this work, the effect of different formulation composition variables on the ASD tablet disintegration performance was systematically studied. GDC-0334: copovidone (PVPVA) 60: 40 ASD prepared by spray drying was selected as the model ASD system. The effects of ASD loading, filler type and ratio, disintegrant type and level were then investigated using tablets made by direct compression process. Tablet disintegration time increased with the increase of ASD loading, especially when ASD loading exceeded 50 %. At the same tablet solid fraction, when lactose was used as the soluble filler, faster tablet disintegration was observed compared to the tablets with mannitol as the soluble filler. Among the three tested disintegrants, croscarmellose sodium performed the best in facilitating the ASD tablet disintegration, followed by sodium starch glycolate, and crospovidone was the poorest. When croscarmellose sodium was used as the disintegrant, 5 % level was sufficient to enable ASD tablet disintegration at 60 % ASD loading and further increase of croscarmellose sodium level to 8 % did not provide additional benefit. Water uptake experiments were performed on selected tablets and the results demonstrated a positive correlation with tablet disintegration time, indicating water penetration is a major contributing step for the disintegration of our ASD tablets. Overall, this work provides a rationale for excipient selection and insights into building a platform formulation approach for developing immediate-release ASD tablets.
Collapse
Affiliation(s)
- Wei Zhang
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Prajwal Thool
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Benjamin W Weitz
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hao Helen Hou
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
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
|
19
|
Indulkar AS, Alex S, Zhang GGZ. Impact of dissolution medium pH and ionization state of the drug on the release performance of amorphous solid dispersions. J Pharm Sci 2025; 114:497-506. [PMID: 39454946 DOI: 10.1016/j.xphs.2024.10.028] [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: 07/31/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Amorphous solid dispersions (ASDs) are widely employed as a strategy to improve oral bioavailability of poorly water soluble compounds. Typically, optimal dissolution performance from a polyvinylpyrrolidone vinyl acetate (PVPVA) based ASD is observed at relatively low drug loading limit. Above a certain drug load, termed limit of congruency (LoC), the release from ASDs significantly decreases. So far, the majority of the dissolution behavior has been tested in conditions where the drug primarily exists in unionized form. In this work, the impact of pH of the dissolution environment on the release performance of ASDs of an ionizable drug was studied. Atazanavir (ATZ), a weakly basic drug with a pKa of 4.5 was used as a model compound and PVPVA was used as a non-ionizable matrix polymer. Dissolution rate was measured using Wood's apparatus which normalizes the surface area of the dissolving tablet. The pH of the dissolution media was varied between 1 and 6.8, to cover a range where ATZ exists as >99 % ionized or unionized species. At pH 6.8, near complete release was observed only when the drug load was ≤ 6 %. Unlike typically observed drastic decline in release behavior for PVPVA based ASDs above LoC, ATZ ASDs underwent gradual decline in dissolution behavior when the DL was increased to 8 %. This was attributed to potential formation of an ATZ-PVPVA associated phase with dissolution rate slower than neat PVPVA. However, the 10 % DL ASD showed negligible ATZ release. On another extreme (pH 1) where ATZ is ∼100 % ionized, the dissolution rate of ATZ was faster than that of PVPVA. ASD dissolution rate was found to be slower than that of the neat drug but faster than PVPVA and interestingly, did not change with DL. This can be attributed to formation of an ionized ATZ-PVPVA phase which controls the dissolution rate of the ASD. At pH 3, where the drug is ∼97 % ionized, near complete release was observed for drug loads ≤ 8 %. To observe significant increase in drug loading with near complete release, >98 % ionization of ATZ was required. At pH 2 where ATZ is ∼99.7 % ionized, near complete release was observed for drug loads up to 30 %. Furthermore, the deterioration in dissolution performance with an increase in drug load continued to be gradual at pH 2. The enhancement in dissolution performance did not correlate with solubility enhancement of ATZ due to ionization. We theorize that the enhancement in the dissolution performance due to ionization is the result of formation of an ionized ATZ-PVPVA phase which increases the hydrophilicity and the miscibility of the ASD. This can help resist water induced phase separation during ASD dissolution and therefore, result in continuous, and congruent dissolution of the drug and polymer.
Collapse
Affiliation(s)
- Anura S Indulkar
- Small Molecule CMC Development, Research and Development, AbbVie Inc., North Chicago, IL 60064, United States.
| | - Samantha Alex
- Small Molecule CMC Development, Research and Development, AbbVie Inc., North Chicago, IL 60064, United States
| | - Geoff G Z Zhang
- Small Molecule CMC Development, Research and Development, AbbVie Inc., North Chicago, IL 60064, United States
| |
Collapse
|
20
|
Hiew TN, Solomos MA, Kafle P, Polyzois H, Zemlyanov DY, Punia A, Smith D, Schenck L, Taylor LS. The importance of surface composition and wettability on the dissolution performance of high drug loading amorphous dispersion formulations. J Pharm Sci 2025; 114:289-303. [PMID: 39349295 DOI: 10.1016/j.xphs.2024.09.020] [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: 06/27/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/02/2024]
Abstract
One of the limitations with an amorphous solid dispersion (ASD) formulation strategy is low drug loading. Hydrophobic drugs have poor wettability and require a substantial amount of polymer to stabilize the amorphous drug and facilitate release. Using grazoprevir and hypromellose acetate succinate as model drug and polymer, respectively, the interplay between particle surface composition, particle wettability, and release performance was investigated. A hierarchical particle approach was used where the surfaces of high drug loading ASDs generated by either solvent evaporation or co-precipitation were further modified with a secondary excipient (i.e., polymer or wetting agent). The surface-modified particles were characterized for drug release, wettability, morphology, and surface composition using two-stage dissolution studies, contact angle measurements, scanning electron microscopy, and X-ray photoelectron spectroscopy, respectively. Despite surface modification with hydrophilic polymers, the hierarchical particles did not consistently exhibit good release performance. Contact angle measurements showed that the secondary excipient had a profound impact on particle wettability. Particles with good wettability showed improved drug release relative to particles that did not wet well, even with similar drug loadings. These observations underscore the intricate interplay between particle wettability and performance in amorphous dispersion formulations and illustrate a promising hierarchical particle approach to formulate high drug loading amorphous dispersions with improved dissolution performance.
Collapse
Affiliation(s)
- Tze Ning Hiew
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Marina A Solomos
- Oral Formulation Sciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Prapti Kafle
- Oral Formulation Sciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Hector Polyzois
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Dmitry Y Zemlyanov
- Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ashish Punia
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Daniel Smith
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Luke Schenck
- Oral Formulation Sciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States.
| |
Collapse
|
21
|
Wdowiak K, Tajber L, Miklaszewski A, Cielecka-Piontek J. Application of the Box-Behnken Design in the Development of Amorphous PVP K30-Phosphatidylcholine Dispersions for the Co-Delivery of Curcumin and Hesperetin Prepared by Hot-Melt Extrusion. Pharmaceutics 2024; 17:26. [PMID: 39861675 PMCID: PMC11768460 DOI: 10.3390/pharmaceutics17010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/20/2024] [Accepted: 12/25/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Curcumin and hesperetin are plant polyphenols known for their poor solubility. To address this limitation, we prepared amorphous PVP K30-phosphatidylcholine dispersions via hot-melt extrusion. Methods: This study aimed to evaluate the effects of the amounts of active ingredients and phosphatidylcholine, as well as the process temperature, on the performance of the dispersions. A Box-Behnken design was employed to assess these factors. Solid-state characterization and biopharmaceutical studies were then conducted. X-ray powder diffraction (XRPD) was used to confirm the amorphous nature of the dispersions, while differential scanning calorimetry (DSC) provided insight into the miscibility of the systems. Fourier-transform infrared spectroscopy (FTIR) was employed to assess the intermolecular interactions. The apparent solubility and dissolution profiles of the systems were studied in phosphate buffer at pH 6.8. In vitro permeability across the gastrointestinal tract and blood-brain barrier was evaluated using the parallel artificial membrane permeability assay. Results: The quantities of polyphenols and phospholipids were identified as significant factors influencing the biopharmaceutical performance of the systems. Solid-state analysis confirmed the formation of amorphous dispersions and the development of interactions among components. Notably, a significant improvement in solubility was observed, with formulations exhibiting distinct release patterns for the active compounds. Furthermore, the in vitro permeability through the gastrointestinal tract and blood-brain barrier was enhanced. Conclusions: The findings suggest that amorphous PVP K30-phosphatidylcholine dispersions have the potential to improve the biopharmaceutical properties of curcumin and hesperetin.
Collapse
Affiliation(s)
- Kamil Wdowiak
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, 3 Rokietnicka St., 60-806 Poznan, Poland;
| | - Lidia Tajber
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, University of Dublin, D02 PN40 Dublin, Ireland;
| | - Andrzej Miklaszewski
- Faculty of Materials Engineering and Technical Physics, Institute of Materials Science and Engineering, Poznan University of Technology, 5 M. Skłodowska-Curie Square, 60-965 Poznan, Poland;
| | - Judyta Cielecka-Piontek
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, 3 Rokietnicka St., 60-806 Poznan, Poland;
| |
Collapse
|
22
|
Tomberg T, Hämäläinen I, Strachan CJ, van Veen B. Dynamic Phase Behavior of Amorphous Solid Dispersions Revealed with In Situ Stimulated Raman Scattering Microscopy. Mol Pharm 2024; 21:6444-6457. [PMID: 39561293 PMCID: PMC11615945 DOI: 10.1021/acs.molpharmaceut.4c01032] [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: 09/11/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/21/2024]
Abstract
This study reports the application of in situ stimulated Raman scattering (SRS) microscopy for real-time chemically specific imaging of dynamic phase phenomena in amorphous solid dispersions (ASDs). Using binary ritonavir and poly(vinylpyrrolidone-vinyl acetate) films with different drug loadings (0-100% w/w) as model systems, we employed SRS microscopy with fast spectral focusing to analyze ASD behavior upon contact with a dissolution medium. Multivariate unmixing of the SRS spectra allowed changes in the distributions of the drug, polymer, and water to be (semi)quantitatively imaged in real time, both in the film and the adjacent dissolution medium. The SRS analyses were further augmented with complementary correlative sum frequency generation and confocal reflection for additional crystallinity and phase sensitivity. In the ASDs with drug loadings of 20, 40, and 60% w/w, the water penetration front within the film, followed by both surface-directed and bulk phase separation in the film, was apparent but differed quantitatively. Additionally, drug-loading and phase-dependent polymer and drug release behavior was imaged, and liquid-liquid phase separation was observed for the 20% drug loading ASD. Overall, SRS microscopy with fast spectral focusing provides quantitative insights into water-induced ASD phase phenomena, with chemical, solid-state, temporal, and spatial resolution. These insights are important for optimal ASD formulation development.
Collapse
Affiliation(s)
- Teemu Tomberg
- Division
of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00790, Finland
| | - Ilona Hämäläinen
- Division
of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00790, Finland
- Pharmaceutical
Sciences, Orion Corporation, Espoo FI-02200, Finland
| | - Clare J. Strachan
- Division
of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki FI-00790, Finland
| | - Bert van Veen
- Pharmaceutical
Sciences, Orion Corporation, Espoo FI-02200, Finland
| |
Collapse
|
23
|
Zhao T, Gu C, Qi J, Liu J, Wang Y, Chen X, Guo F, Li Y. In vitro and in vivo performance of amorphous solid dispersions of ursolic acid as a function of polymer type and excipient addition. J Pharm Pharmacol 2024; 76:1584-1598. [PMID: 39393786 DOI: 10.1093/jpp/rgae125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/14/2024] [Indexed: 10/13/2024]
Abstract
OBJECTIVES The objective of this research was to enhance the bioavailability of ursolic acid (UA) by preparing multielement amorphous solid dispersion (ASD) systems comprising excipients. METHODS The ASDs were prepared via the solvent evaporation method, characterized by a range of techniques, and investigated with respect to permeability of human colorectal adenocarcinoma cell line (Caco-2) cells monolayers and pharmacokinetics, with comparisons made to the physical mixture and the pure drug. KEY FINDINGS The (UA-choline)-Polyethylcaprolactam-polyvinyl acetate-polyethylene glycol grafted copolymer (Soluplus)-Vitamin E polyethylene glycol succinate (TPGS) ASD demonstrated superior dissolution properties compared to the corresponding binary solid dispersions and ternary solid dispersions (P< .05). The permeability studies of Caco-2 cell monolayers revealed that the ASD exhibited moderate permeability, with an efflux rate that was significantly lower than that of the UA raw material (P< .05). Pharmacokinetic studies in rats demonstrated that the oral bioavailability of the ASD was 19.0 times higher than that of UA (P< .01). CONCLUSIONS The research indicated that the multielement ASD could be employed as an efficacious drug delivery system for UA. Furthermore, the Soluplus/TPGS/choline combination represents a promising candidate for the fabrication of ASDs that can load weakly acidic and poorly soluble drugs.
Collapse
Affiliation(s)
- Tingting Zhao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, People's Republic of China
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, 369 Tianxiong Road, Shanghai 201318, People's Republic of China
| | - Chenming Gu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, People's Republic of China
| | - Jianbo Qi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, People's Republic of China
| | - Jingwen Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, People's Republic of China
| | - Yajun Wang
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, 369 Tianxiong Road, Shanghai 201318, People's Republic of China
| | - Xiaojing Chen
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, 369 Tianxiong Road, Shanghai 201318, People's Republic of China
| | - Fujiang Guo
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, People's Republic of China
| | - Yiming Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, People's Republic of China
| |
Collapse
|
24
|
Bharti K, Jha A, Kumar M, Manjit, Satpute AP, Akhilesh, Tiwari V, Mishra B. Correlation of surface properties with dissolution behavior of amorphous solid dispersion of Riluzole and its pharmacodynamic evaluation. J Pharm Sci 2024; 113:3554-3564. [PMID: 39414079 DOI: 10.1016/j.xphs.2024.10.010] [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: 05/31/2024] [Revised: 10/05/2024] [Accepted: 10/05/2024] [Indexed: 10/18/2024]
Abstract
Formulation of amorphous solid dispersion (ASD) of any poorly water-soluble drug is among the most promising techniques to increase the dissolution profile of drug and hence its bioavailability. Various literatures give evidences of the role of drug-polymer interactions in the ASD systems, very little information is available about the surface properties of the drug molecule and their ASDs which contributes to a higher dissolution profile. Current work focuses on exploring the surface behavior of a poorly water-soluble drug Riluzole (RLZ) and its ASDs prepared with two highly hydrophilic polymers, polyacrylic acid (PAA), and polyvinylpyrrolidone vinyl acetate (PVP VA). Initial characterization using X-ray diffraction (XRD) revealed about the weight fraction of drug required to prepare a single-phase homogenous system with both the polymers. The saturation solubility and the dissolution studies showed an increase in RLZ solubility as well as the dissolution profile due to the presence of polymers. The role of polymers in changing the surface properties in terms of wettability and polarity were explored using contact angle method and X-ray photon spectroscopy (XPS). Additionally, the neuroprotective efficacy and dose dependent hepatotoxicity were also evaluated in male wistar rats. These studies confirmed the increase in the surface polarity and hence the enhanced ability of ASD formulations to interact with water. The in vivo studies indicated that at the current recommended dose the efficacy as well as toxicity is increased for the ASD formulation. Hence, this formulation can be given at a lower dose to achieve same therapeutic effect with lower toxicity.
Collapse
Affiliation(s)
- Kanchan Bharti
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, U.P. 221005, India.
| | - Abhishek Jha
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, U.P. 221005, India.
| | - Manish Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, U.P. 221005, India.
| | - Manjit
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, U.P. 221005, India.
| | - Amol Parasram Satpute
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, U.P. 221005, India.
| | - Akhilesh
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, U.P. 221005, India.
| | - Vinod Tiwari
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, U.P. 221005, India.
| | - Brahmeshwar Mishra
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, U.P. 221005, India.
| |
Collapse
|
25
|
Men S, Polli JE. Microscope-enabled disc dissolution system: Concordance between drug and polymer dissolution from an amorphous solid dispersion disc and visual disc degradation. J Pharm Sci 2024; 113:3586-3598. [PMID: 39454944 DOI: 10.1016/j.xphs.2024.10.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/20/2024] [Accepted: 10/20/2024] [Indexed: 10/28/2024]
Abstract
A microscopic erosion time test was recently described to anticipate amorphous solid dispersion (ASD) drug load dispersibility limit, using 0.5 ml media volume. Studies here build upon this microscope-enabled method but focus on drug and polymer dissolution from an ASD disc, along with imaging. The objective was 1) to design and build a microscope-enabled disc dissolution system (MeDDiS) with a 900 mL dissolution volume and 2) assess the ability of MeDDiS imaging of dissolving discs to provide concordance with measured drug and polymer dissolution profiles. MeDDiS employed a digital microscope to image ASD discs and a one-liter vessel for dissolution. ASD discs containing ritonavir (5-50 % drug load) and PVPVA were fabricated and subjected to in vitro dissolution using MeDDiS, where disc diameter was quantified with time. Ritonavir and PVPVA release were also measured. Results indicate concordance between imaging and dissolution. Both found 25 % drug load to provide high drug and polymer release, but 30 % yielded low release. Quantitatively, MeDDiS images predicted drug and polymer release profiles, both above and below the drug load cliff. Overall, studies here describe a MeDDiS which has promised to anticipate drug and polymer dissolution, via imaging of dissolving discs, above and below the ASD drug load cliff.
Collapse
Affiliation(s)
- Shuaiqian Men
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - James E Polli
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA.
| |
Collapse
|
26
|
Oktay AN, Polli JE. Screening of Polymers for Oral Ritonavir Amorphous Solid Dispersions by Film Casting. Pharmaceutics 2024; 16:1373. [PMID: 39598497 PMCID: PMC11597764 DOI: 10.3390/pharmaceutics16111373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/17/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Drug-polymer interactions and miscibility promote the formation and performance of amorphous solid dispersions (ASDs) of poorly soluble drugs for improved oral bioavailability. The objective of this study was to employ drug-polymer interaction calculations and small-scale experimental characterization to screen polymers for potential ASDs of ritonavir. Methods: Seven polymers across four polymer types were screened as follows: an enteric one (EudragitS100), amphiphilic ones (HPMCAS-L, HPMCAS-H, and their 1:1 combination), hydrophilic ones (PEG-6000, PVP-VA), and a surfactant (Soluplus), including PVP-VA as a positive control, as the commercial ASD employs PVP-VA. Drug-polymer interaction calculations were performed for Hansen solubility parameter, Flory-Huggins parameter, and glass transition temperature. ASDs were prepared via film casting. Experimental characterizations included drug solubility in polymer solutions, polymer inhibition of drug precipitation, polarized light microscopy, differential scanning calorimetry, solubilization capacity, and dissolution studies. Results: HPMCAS-L, HPMCAS L:H, and Soluplus, along with the positive control PVP-VA, were identified as polymers for potential ASDs of ritonavir, with HPMCAS-L and PVP-VA being preferable. HPMCAS-L and the positive control PVP-VA were always viable for both 20% and 40% drug loads across all tests. Films with each of these four polymers showed improved dissolution compared to amorphous ritonavir without polymer. Drug-polymer interaction calculations anticipated the unfavorable small-scale experimental results for PEG-6000 and EudragitS100. Conclusion: Overall, the results contribute towards a resource-sparing approach to identify polymers for ASDs.
Collapse
Affiliation(s)
- Ayse Nur Oktay
- Department of Pharmaceutical Technology, Gulhane Faculty of Pharmacy, University of Health Sciences, Ankara 06018, Türkiye
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD 21201, USA
| | - James E. Polli
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD 21201, USA
| |
Collapse
|
27
|
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
|
28
|
Walter S, Mileo PGM, Afzal MAF, Kyeremateng SO, Degenhardt M, Browning AR, Shelley JC. Predicting the Release Mechanism of Amorphous Solid Dispersions: A Combination of Thermodynamic Modeling and In Silico Molecular Simulation. Pharmaceutics 2024; 16:1292. [PMID: 39458621 PMCID: PMC11510624 DOI: 10.3390/pharmaceutics16101292] [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: 08/14/2024] [Revised: 09/18/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND During the dissolution of amorphous solid dispersion (ASD) formulations, the drug load (DL) often impacts the release mechanism and the occurrence of loss of release (LoR). The ASD/water interfacial gel layer and its specific phase behavior in connection with DL strongly dictate the release mechanism and LoR of ASDs, as reported in the literature. Thermodynamically driven liquid-liquid phase separation (LLPS) and/or drug crystallization at the interface are the key phase transformations that drive LoR. METHODS In this study, a combination of Perturbed-Chain Statistical Associating Fluid Theory (PC-SAFT) thermodynamic modeling and in silico molecular simulation was applied to investigate the release mechanism and the occurrence LoR of an ASD formulation consisting of ritonavir as the active pharmaceutical ingredient (API) and the polymer, polyvinylpyrrolidone-co-vinyl acetate (PVPVA64). A thermodynamically modeled ternary phase diagram of ritonavir (PVPVA64) and water was applied to predict DL-dependent LLPS in the ASD/water interfacial gel layer. Microscopic Erosion Time Testing (METT) was used to experimentally validate the phase diagram predictions. Additionally, in silico molecular simulation was applied to provide further insights into the phase separation, the release mechanism, and aggregation behavior on a molecular level. RESULTS Thermodynamic modeling, molecular simulation, and experimental results were consistent and complementary, providing evidence that ASD/water interactions and phase separation are essential factors driving the dissolution behavior and LoR at 40 wt% DL of the investigated ritonavir/PVPVA64 ASD system, consistent with previous studies. CONCLUSIONS This study provides insights into the potential of blending thermodynamic modeling, molecular simulation, and experimental research to comprehensively understand ASD formulations. Such a combined approach can be leveraged as a computational framework to gain insights into the ASD dissolution mechanism, thereby facilitating in silico screening, designing, and optimization of formulations with the benefit of significantly reducing the number of experimental tests.
Collapse
Affiliation(s)
- Stefanie Walter
- AbbVie Deutschland GmbH & Co. KG, Small Molecule CMC Development, Knollstraße, 67061 Ludwigshafen am Rhein, Germany; (S.W.); (M.D.)
| | - Paulo G. M. Mileo
- Materials Science, Schrödinger GmbH, Glücksteinallee 25, 68163 Mannheim, Germany;
| | - Mohammad Atif Faiz Afzal
- Materials Science, Schrödinger LLC, Suite 1300, 101 SW Main Street, Portland, OR 97204, USA; (M.A.F.A.); (A.R.B.)
| | - Samuel O. Kyeremateng
- AbbVie Deutschland GmbH & Co. KG, Small Molecule CMC Development, Knollstraße, 67061 Ludwigshafen am Rhein, Germany; (S.W.); (M.D.)
| | - Matthias Degenhardt
- AbbVie Deutschland GmbH & Co. KG, Small Molecule CMC Development, Knollstraße, 67061 Ludwigshafen am Rhein, Germany; (S.W.); (M.D.)
| | - Andrea R. Browning
- Materials Science, Schrödinger LLC, Suite 1300, 101 SW Main Street, Portland, OR 97204, USA; (M.A.F.A.); (A.R.B.)
| | - John C. Shelley
- Life Science, Schrödinger LLC, Suite 1300, 101 SW Main Street, Portland, OR 97204, USA
| |
Collapse
|
29
|
Zhuo X, Jasiukenaite I, Löbmann K. β-Lactoglobulin-based amorphous solid dispersions: A graphical review on the state-of-the-art. Eur J Pharm Biopharm 2024; 202:114396. [PMID: 38971201 DOI: 10.1016/j.ejpb.2024.114396] [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: 05/16/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Proteins have recently caught attention as potential excipients for amorphous solid dispersions (ASDs) to improve oral bioavailability of poorly water-soluble drugs. Notably, the studies have highlighted whey protein isolates, particularly β-lactoglobulin (BLG), as promising candidates in amorphous stabilization, dissolution and solubility enhancement, achieving drug loadings of 50 wt% and higher. Consequently, investigations into the mechanisms underlying the solid-state stabilization of amorphous drugs and the enhancement of drug solubility in solution have been conducted. This graphical review provides a comprehensive overview of recent findings concerning BLG-based ASDs. Firstly, the dissolution performance of BLG-based ASDs is compared to more traditional polymer-based ASDs. Secondly, the drug loading onto BLG and the resulting amorphous stabilization mechanisms is summarized. Thirdly, interactions between BLG and drug molecules in solution are described as the mechanisms governing the improvement of drug solubility. Lastly, we outline the impact of the spray drying process on the secondary structure of BLG, and the resulting differences in amorphous stabilization and drug dissolution performance between α-helix-rich and β-sheet-rich BLG-based ASDs.
Collapse
Affiliation(s)
- Xuezhi Zhuo
- Zerion Pharma A/S, Fruebjergvej 3, 2100 Copenhagen, Denmark
| | | | | |
Collapse
|
30
|
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
|
31
|
Barr KE, Ohnsorg ML, Liberman L, Corcoran LG, Sarode A, Nagapudi K, Feder CR, Bates FS, Reineke TM. Drug-Polymer Nanodroplet Formation and Morphology Drive Solubility Enhancement of GDC-0810. Bioconjug Chem 2024; 35:499-516. [PMID: 38546823 DOI: 10.1021/acs.bioconjchem.4c00018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2024]
Abstract
Nanodroplet formation is important to achieve supersaturation of active pharmaceutical ingredients (APIs) in an amorphous solid dispersion. The aim of the current study was to explore how polymer composition, architecture, molar mass, and surfactant concentration affect polymer-drug nanodroplet morphology with the breast cancer API, GDC-0810. The impact of nanodroplet size and morphology on dissolution efficacy and drug loading capacity was explored using polarized light microscopy, dynamic light scattering, and cryogenic transmission electron microscopy. Poly(N-isopropylacrylamide-stat-N,N-dimethylacrylamide) (PND) was synthesized as two linear derivatives and two bottlebrush derivatives with carboxylated or PEGylated end-groups. Hydroxypropyl methylcellulose acetate succinate grade MF (HPMCAS-MF) and poly(vinylpyrrolidone-co-vinyl acetate) (PVPVA) were included as commercial polymer controls. We report the first copolymerization synthesis of a PVPVA bottlebrush copolymer, which was the highest performing excipient in this study, maintaining 688 μg/mL GDC-0810 concentration at 60 wt % drug loading. This is likely due to strong polymer-drug noncovalent interactions and the compaction of GDC-0810 along the PVPVA bottlebrush backbone. Overall, it was observed that the most effective formulations had a hydrodynamic radius less than 25 nm with tightly compacted nanodroplet morphologies.
Collapse
Affiliation(s)
- Kaylee E Barr
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Monica L Ohnsorg
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Lucy Liberman
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Louis G Corcoran
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Apoorva Sarode
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, California 94080, United States
| | - Karthik Nagapudi
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, California 94080, United States
| | - Christina R Feder
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, California 94080, United States
| | - Frank S Bates
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
32
|
Purohit HS, Zhou D, Yu M, Zaroudi M, Oberoi H, López ADLR, Kelkar MS, He Y, Gates B, Nere N, Law D. Proof-of-Concept in Developing a 45% Drug Loaded Amorphous Nanoparticle Formulation. J Pharm Sci 2024; 113:1007-1019. [PMID: 37832919 DOI: 10.1016/j.xphs.2023.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023]
Abstract
Amorphous solid dispersion (ASD) is an enabling approach utilized to deliver poorly soluble compounds. ASDs can spontaneously generate drug-rich amorphous nanoparticles upon dissolution, which can act as a reservoir for maintaining supersaturation during oral absorption. But, conventional ASDs are often limited in drug loadings to < 20 %. For indications where the dose is high, this can translate into a significant pill burden. The aim of this research was to develop a high drug loading (DL) amorphous nanoparticle (ANP) formulation that can release the drug-rich nanoparticles into solution upon contact with aqueous environment. Nanoparticles were directly engineered using solvent/anti-solvent precipitation. The obtained nanoparticle suspension was then concentrated followed by solidification to a re-dispersible amorphous dosage form using spray drying or lyophilization. The impact of process variables was studied using dynamic light scattering (DLS), scanning electron microscopy (SEM), high performance liquid chromatography (HPLC), nuclear magnetic resonance (NMR) and differential scanning calorimetry (DSC). It was observed that spray drying led to a non-re-dispersible formulation. Sucrose and trehalose containing lyocakes resulted in re-dispersible formulations. The trehalose containing lyocakes, in a dog study, gave comparable performance to the reference tablet in the fasted state but lower area under the curve (AUC) in fed state.
Collapse
Affiliation(s)
- Hitesh S Purohit
- Small molecule CMC development, Drug Product Development, AbbVie Inc., North Chicago, IL, USA.
| | - Deliang Zhou
- Small Molecule Drug Product Development, BeiGene, Beijing, China
| | - Mengqi Yu
- Small molecule CMC development, Drug Product Development, AbbVie Inc., North Chicago, IL, USA
| | | | - Hardeep Oberoi
- Small molecule CMC development, Drug Product Development, AbbVie Inc., North Chicago, IL, USA
| | | | - Manish S Kelkar
- Small molecule CMC development, Process Engineering, AbbVie Inc., North Chicago, IL, USA
| | - Yan He
- Small molecule CMC development, Analytical Research and Development, AbbVie Inc., North Chicago, IL, USA
| | - Bradley Gates
- Small molecule CMC development, Process Chemistry, AbbVie Inc., North Chicago, IL, USA
| | - Nandkishor Nere
- Small molecule CMC development, Process Engineering, AbbVie Inc., North Chicago, IL, USA
| | - Devalina Law
- Small molecule CMC development, Drug Product Development, AbbVie Inc., North Chicago, IL, USA.
| |
Collapse
|
33
|
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
|
34
|
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
|
35
|
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
|
36
|
Yu D, Nie H, Hoag SW. Comprehensive evaluation of polymer types and ratios in Spray-Dried Dispersions: Compaction, Dissolution, and physical stability. Int J Pharm 2024; 650:123674. [PMID: 38061497 DOI: 10.1016/j.ijpharm.2023.123674] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 11/13/2023] [Accepted: 12/03/2023] [Indexed: 01/08/2024]
Abstract
Amorphous solid dispersion (ASD) is a well-established strategy for enhancing the solubility and bioavailability of poorly soluble drugs. A significant portion of ASD products are in tablet form. However, the influence of common polymers and drug loading on the manufacturability of ASD tablets remains underexplored. This study focuses on investigating spray-dried ASDs from a tableting perspective by evaluating their physiochemical and mechanical properties. Itraconazole (ITZ) and indomethacin (IND), at the drug loadings ranging from 10% to 50%, were prepared with two polymers, hydroxypropyl methylcellulose acetate succinate (HPMCAS) and polyvinylpyrrolidone (PVP), serving as representative systems. Our findings revealed that increasing the drug loading resulted in a decreased surface area in ITZ-HPMCAS, IND-HPMCAS, and IND-PVP ASDs. However, this trend was not observed in ITZ-PVP dispersions, possibly due to the morphological disparities. Compaction results demonstrated that tabletability improved with decreasing drug loadings, except for ITZ-PVP dispersions. A partial least square analysis underscored particle surface area as the key factor influencing the tensile strength of ASD tablets. Additionally, our study disclosed that ITZ-PVP ASDs exhibited the worst release profiles and stability performance. The comprehensive journey from characterizing ASD particles to analyzing their compaction behavior and investigating drug release and physical stability offered profound insights into the attributes crucial for the downstream processing of amorphous pharmaceuticals.
Collapse
Affiliation(s)
- Dongyue Yu
- University of Maryland, Baltimore, School of Pharmacy, Baltimore, MD 21201, United States
| | - Haichen Nie
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, United States
| | - Stephen W Hoag
- University of Maryland, Baltimore, School of Pharmacy, Baltimore, MD 21201, United States.
| |
Collapse
|
37
|
Handa U, Malik A, Guarve K, Rani N, Sharma P. Supersaturation Behavior: Investigation of Polymers Impact on Nucleation Kinetic Profile for Rationalizing the Polymeric Precipitation Inhibitors. Curr Drug Deliv 2024; 21:1422-1432. [PMID: 37907490 DOI: 10.2174/0115672018261505231018100329] [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: 05/09/2023] [Revised: 07/26/2023] [Accepted: 09/01/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND Although nucleation kinetic data is quite important for the concept of supersaturation behavior, its part in rationalizing the crystallization inhibitor has not been well understood. OBJECTIVE This study aimed to investigate the nucleation kinetic profile of Dextromethorphan HBr (as an ideal drug, BCS-II) by measuring liquid-liquid phase segregation, nucleation induction time, and Metastable Zone width. METHODS Surfeit action was examined by a superfluity assay of the drug. The concentration was scrutinized by light scattering techniques (UV spectrum (novel method) and Fluorometer (CL 53)). RESULTS The drug induction time was 20 min without polymer and 90 and 110 min with polymers, such as HPMC K15M and Xanthan Gum, respectively. Therefore, the order of the polymer's ability to inhibit nucleation was Xanthan Gum > HPMC K15M in the medium (7.4 pH). Similarly, the drug induction time was 30 min without polymer and 20, 110, and 90 min with polymers, such as Sodium CMC, HPMC K15M, and Xanthan Gum, respectively. Therefore, the order of the polymer's ability to inhibit nucleation was HPMC K15M > Xanthan Gum > Sodium CMC in SIFsp (6.8 pH), which synchronizes the polymer's potentiality to interdict the drug precipitation. CONCLUSION The HPMC K15M and xanthan Gum showed the best crystallization inhibitor effect for the maintenance of superfluity conditions till the drug absorption time. The xanthan gum is based on the "glider" concept, and this shows the novelty of this preliminary research. The screening methodology used for rationalizing the best polymers used in the superfluity formulations development successfully.
Collapse
Affiliation(s)
- Uditi Handa
- Guru Gobind Singh College of Pharmacy, Yamuna Nagar-135001, Haryana, India
- Department of Pharmaceutics, MM College of Pharmacy, MM (DU), Mullana, Ambala, Haryana, India
| | - Anuj Malik
- Department of Pharmaceutics, MM College of Pharmacy, MM (DU), Mullana, Ambala, Haryana, India
| | - Kumar Guarve
- Guru Gobind Singh College of Pharmacy, Yamuna Nagar-135001, Haryana, India
| | - Nidhi Rani
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Prerna Sharma
- Guru Gobind Singh College of Pharmacy, Yamuna Nagar-135001, Haryana, India
| |
Collapse
|
38
|
Huzjak T, Jakasanovski O, Berginc K, Puž V, Zajc-Kreft K, Jeraj Ž, Janković B. Overcoming drug impurity challenges in amorphous solid dispersion with rational development of biorelevant dissolution-permeation method. Eur J Pharm Sci 2024; 192:106655. [PMID: 38016626 DOI: 10.1016/j.ejps.2023.106655] [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: 08/01/2023] [Revised: 10/26/2023] [Accepted: 11/25/2023] [Indexed: 11/30/2023]
Abstract
Hot-melt extrusion is often used to prepare amorphous solid dispersion to overcome low drug solubility and enhance bio-performance of the formulation. Due to the uniqueness of each drug - polymer combination and its physico-chemical properties, setting the appropriate HME barrel temperature, feed rate and screw speed ensures drug amorphization, absence of residual crystallinity, absence of water, and a suitable drug release profile. In this research, samples with BCS II/IV model drug and PVP/VA polymer were prepared to evaluate the impact of HME process parameters, incoming drug form (anhydrous vs. hydrate), and drug supplier (i.e., impurity profile), on biorelevant drug release. This study provides a relationship between observed in vitro supersaturation and precipitation behavior of amorphous solid dispersion formulation with in vivo results, on patients, by using the acceptor profile of side-by-side dissolution-permeation apparatus. An in vitro dissolution method, in small volumes, in an apparatus with paddles and dissolution-permeation side-by-side method was developed on the MicroFlux™ apparatus to assess if the differences observed in vitro bears relevance to the bioequivalence outcome in vivo. The former was used to guide the generic drug product development due to high discriminatory strength, while the latter was biorelevant, due to the inclusion of the second compartment assuring absorptive environment to capture the impact of supersaturation and subsequent precipitation on bioavailability. Bio-relevancy of the in vitro method was confirmed with the in vivo dog study and clinical study on patients, and an in vitro - in vivo correlation was established. For the investigated BCS II/IV drug, this research highlights the importance of considering supersaturation and formation of colloidal species during amorphous solid dispersion release testing to assure product quality, safety and efficacy.
Collapse
Affiliation(s)
- T Huzjak
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, Ljubljana 1000, Slovenia; Product Development, Lek Pharmaceuticals d.d., Verovškova 57, Ljubljana 1526, Slovenia.
| | - O Jakasanovski
- Product Development, Lek Pharmaceuticals d.d., Verovškova 57, Ljubljana 1526, Slovenia
| | - K Berginc
- Product Development, Lek Pharmaceuticals d.d., Verovškova 57, Ljubljana 1526, Slovenia
| | - V Puž
- Product Development, Lek Pharmaceuticals d.d., Verovškova 57, Ljubljana 1526, Slovenia
| | - K Zajc-Kreft
- Product Development, Lek Pharmaceuticals d.d., Verovškova 57, Ljubljana 1526, Slovenia
| | - Ž Jeraj
- Product Development, Lek Pharmaceuticals d.d., Verovškova 57, Ljubljana 1526, Slovenia
| | - B Janković
- Product Development, Lek Pharmaceuticals d.d., Verovškova 57, Ljubljana 1526, Slovenia
| |
Collapse
|
39
|
Li J, Wang X, Yu D, Zhoujin Y, Wang K. Molecular complexes of drug combinations: A review of cocrystals, salts, coamorphous systems and amorphous solid dispersions. Int J Pharm 2023; 648:123555. [PMID: 37890646 DOI: 10.1016/j.ijpharm.2023.123555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/17/2023] [Accepted: 10/23/2023] [Indexed: 10/29/2023]
Abstract
As the advancements in the medical technology and healthcare develop through the years, combinational therapy has evolved to be an important treatment modality in many disease settings, including cancer, cardiovascular disease and infectious diseases. In an effort to alleviate "pill burden" and improve patient compliance, fixed dose combinations (FDCs) have been developed to be used as effective therapeutics. Among all FDCs, the category of drug-drug molecular complexes has been proven an efficient methodology in designing and treating diseases, with many drugs being approved. Among all drug-drug molecular complexes, drug-drug cocrystals, salts, coamorphous systems and solid dispersions have been successfully developed and many have been approved by the FDA. In this review, we dwell deeply into the molecular mechanisms behind the different types of drug-drug molecular complexes, including the key functional groups involved in the intermolecular interactions, the applications of each category of molecular complexes, as well as the advantages and challenges thereof. This comprehensive review provides useful insights into the practical design and manufacture of drug-drug molecular complexes and points out the future direction for the development of new advantageous combinational therapies that benefit more patients.
Collapse
Affiliation(s)
- Jinghan Li
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Xiyan Wang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | - Dongyue Yu
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, NJ 08540, United States
| | - Yunping Zhoujin
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, United States
| | - Kunlin Wang
- BeBetter Med Inc., Guangzhou, 510663, PR China; College of Pharmacy, Jinan University, Guangzhou, 510006, PR China.
| |
Collapse
|
40
|
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
|
41
|
Mamidi H, Palekar S, Patel H, Nukala PK, Patel K. Formulation strategies for the development of high drug-loaded amorphous solid dispersions. Drug Discov Today 2023; 28:103806. [PMID: 37890714 DOI: 10.1016/j.drudis.2023.103806] [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: 07/11/2023] [Revised: 10/15/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023]
Abstract
Amorphous solid dispersions (ASD) have gained tremendous attention over the past two decades as one of the most promising techniques for enhancing the solubility of poorly water-soluble drugs. However, low drug loading is one of the major challenges of ASD technology that limits its commercialization to only a few drug candidates. Increasing the drug loading increases the risk of recrystallization during storage (solid state) and/or during dissolution (solution state). Various formulation and process-related strategies have been explored that open the possibility of formulating high drug-loaded ASDs without the risk of recrystallization. Here, we review various formulation approaches, such as the use of surfactants, mesoporous silicas, polymer combinations, in situ thermal crosslinking, structural modification of polymeric carriers, and surface nanocoating using minerals. We also discuss the mechanisms by which these approaches inhibit solid state and/or solution state recrystallization.
Collapse
Affiliation(s)
- Hemanth Mamidi
- College of Pharmacy and Health Sciences, St John's University, Queens, NY, USA; Continuus Pharmaceuticals Inc, Woburn, MA, USA
| | - Siddhant Palekar
- College of Pharmacy and Health Sciences, St John's University, Queens, NY, USA
| | - Henis Patel
- College of Pharmacy and Health Sciences, St John's University, Queens, NY, USA
| | - Pavan Kumar Nukala
- College of Pharmacy and Health Sciences, St John's University, Queens, NY, USA
| | - Ketan Patel
- College of Pharmacy and Health Sciences, St John's University, Queens, NY, USA.
| |
Collapse
|
42
|
Yang R, Zhang GGZ, Zemlyanov DY, Purohit HS, Taylor LS. Drug Release from Surfactant-Containing Amorphous Solid Dispersions: Mechanism and Role of Surfactant in Release Enhancement. Pharm Res 2023; 40:2817-2845. [PMID: 37052841 DOI: 10.1007/s11095-023-03502-3] [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: 01/06/2023] [Accepted: 03/18/2023] [Indexed: 04/14/2023]
Abstract
PURPOSE To understand how surfactants affect drug release from ternary amorphous solid dispersions (ASDs), and to investigate different mechanisms of release enhancement. METHODS Ternary ASDs containing ritonavir (RTV), polyvinylpyrrolidone/vinyl acetate (PVPVA) and a surfactant (sodium dodecyl sulfate (SDS), Tween 80, Span 20 or Span 85) were prepared with rotary evaporation. Release profiles of ternary ASDs were measured with surface normalized dissolution. Phase separation morphologies of ASD compacts during hydration/dissolution were examined in real-time with a newly developed confocal fluorescence microscopy method. The water ingress rate of different formulations was measured with dynamic vapor sorption. Microscopy was employed to check for matrix crystallization during release studies. RESULTS All surfactants improved drug release at 30% DL, while only SDS and Tween 80 improved drug release at higher DLs, although SDS promoted matrix crystallization. The dissolution rate of neat polymer increased when SDS and Tween 80 were present. The water ingress rate also increased in the presence of all surfactants. Surfactant-incorporation affected both the kinetic and thermodynamics factors governing phase separation of RTV-PVPVA-water system, modifying the phase morphology during ASD dissolution. Importantly, SDS increased the miscibility of RTV-PVPVA-water system, whereas other surfactants mainly affected the phase separation kinetics/drug-rich barrier persistence. CONCLUSION Incorporation of surfactants enhanced drug release from RTV-PVPVA ASDs compared to the binary system. Increased drug-polymer-water miscibility and disruption of the drug-rich barrier at the gel-solvent interface via plasticization are highlighted as two key mechanisms underlying surfactant impacts based on direct visualization of the phase separation process upon hydration and release.
Collapse
Affiliation(s)
- Ruochen Yang
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
| | - Geoff G Z Zhang
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, IL, 60064, USA
| | - Dmitry Y Zemlyanov
- Birck Nanotechnology Center, Purdue University, West Lafayette, IN, 47907, USA
| | - Hitesh S Purohit
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, IL, 60064, USA.
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
43
|
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
|
44
|
Yu M, Oberoi HS, Purohit HS, Fowler CA, Law D. Design of Redispersible High-Drug-Load Amorphous Formulations: Impact of Ionic vs Nonionic Surfactants on Processing and Performance. Mol Pharm 2023; 20:5827-5841. [PMID: 37876176 DOI: 10.1021/acs.molpharmaceut.3c00684] [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] [Indexed: 10/26/2023]
Abstract
Amorphous solid dispersions (ASDs) are an enabling formulation approach used to enhance bioavailability of poorly water-soluble molecules in oral drug products. Drug-rich amorphous nanoparticles generated in situ during ASD dissolution maintain supersaturation that drives enhanced absorption. However, in situ formation of nanoparticles requires large quantities of polymers to release drugs rapidly, resulting in an ASD drug load <25%. Delivering directly engineered drug-rich amorphous nanoparticles can reduce the quantities of polymers significantly without sacrificing bioavailability. Preparation of 90% drug-load amorphous nanoparticles (ANPs) of <300 nm diameter using solvent/antisolvent nanoprecipitation, organic solvent removal, and spray drying was demonstrated previously on model compound ABT-530 with Copovidone and sodium dodecyl sulfate (anionic). In this work, nonionic surfactant d-α-tocopheryl polyethylene glycol succinate (Vitamin E TPGS, or TPGS) was used to prepare ANPs as a comparison. Characterization of ANPs by dynamic light scattering, filtrate potency assay, scanning electron microscopy, and differential scanning calorimetry revealed differences in surface properties of nanoparticles afforded by surfactants. This work demonstrates the importance of understanding the impact of the stabilizing agents on nanoparticle behavior when designing a high-drug-load amorphous formulation for poorly water-soluble compounds as well as the impact on redispersion.
Collapse
Affiliation(s)
- Mengqi Yu
- Research & Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Hardeep S Oberoi
- Research & Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Hitesh S Purohit
- Research & Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Craig A Fowler
- Research & Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Devalina Law
- Research & Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| |
Collapse
|
45
|
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
|
46
|
Nunes PD, Pinto JF, Bauer-Brandl A, Brandl M, Henriques J, Paiva AM. In vitro dissolution/permeation tools for amorphous solid dispersions bioavailability forecasting I: Experimental design for PermeaLoop™. Eur J Pharm Sci 2023; 188:106512. [PMID: 37423576 DOI: 10.1016/j.ejps.2023.106512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/11/2023] [Accepted: 06/29/2023] [Indexed: 07/11/2023]
Abstract
Along with the increasing demand for candidate-enabling formulations comes the need for appropriate in vitro bioavailability forecasting. Dissolution/permeation (D/P) systems employing cell-free permeation barriers are increasingly gaining interest, due to their low cost and easy application as passive diffusion bio-predictive profiling in drug product development, as this accounts for nearly 75% of new chemical entities (NCEs) absorption mechanism. To this end, this study comprises theoretical considerations on the design and experimental work towards the establishment and optimization of a PermeaLoop™ based dissolution/permeation assay to simultaneously evaluate the drug release and permeation using Itraconazole (ITZ)-based amorphous solid dispersions (ASD) formulations, with different drug loads, based on a solvent-shift approach. Alternative method conditions were tested such as: donor medium, acceptor medium and permeation barrier were screened using both PermeaPad® and PermeaPlain® 96-well plates. A range of solubilizers, namely Sodium Dodecyl Sulfate, Vitamin E-TPGS and hydroxypropyl-β-cyclodextrin, were screened as possible solubilizing additives to the acceptor medium, while donor medium was varied between blank FaSSIF (phosphate buffer) and FaSSIF. The method optimization also included the ITZ dose selection, being the ITZ single dose (100 mg) considered the most adequate to be used in further experiments to allow the comparison with in vivo studies. In the end, a standardized approach that may be applied to predict the bioavailability of weakly basic poorly soluble drug-based formulations is described, contributing to strengthening the analytical portfolio of in vitro pre-clinical drug product development.
Collapse
Affiliation(s)
- Patrícia D Nunes
- R&D Analytical Development, Hovione Farmaciência S.A., Campus do Lumiar, Building S, 1649-038 Lisboa, Portugal; R&D Oral Drug Product Development, Hovione Farmaciência S.A., Campus do Lumiar, Building S, 1649-038 Lisboa, Portugal; Research Institute for Medicines (iMed.Ulisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - João F Pinto
- Research Institute for Medicines (iMed.Ulisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal.
| | - Annette Bauer-Brandl
- Drug Transport and Delivery Group, Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense DK, 5230, Denmark
| | - Martin Brandl
- Drug Transport and Delivery Group, Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense DK, 5230, Denmark.
| | - João Henriques
- R&D Oral Drug Product Development, Hovione Farmaciência S.A., Campus do Lumiar, Building S, 1649-038 Lisboa, Portugal
| | - Ana Mafalda Paiva
- R&D Analytical Development, Hovione Farmaciência S.A., Campus do Lumiar, Building S, 1649-038 Lisboa, Portugal
| |
Collapse
|
47
|
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
|
48
|
Nunes PD, Ferreira AF, Pinto JF, Bauer-Brandl A, Brandl M, Henriques J, Paiva AM. In vitro dissolution/permeation tools for amorphous solid dispersions bioavailability forecasting II: Comparison and mechanistic insights. Eur J Pharm Sci 2023; 188:106513. [PMID: 37423577 DOI: 10.1016/j.ejps.2023.106513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/11/2023] [Accepted: 06/29/2023] [Indexed: 07/11/2023]
Abstract
Along with the increasing demand for complex formulations comes the need for appropriate in vitro methodologies capable of predicting their corresponding in vivo performance and the mechanisms controlling the drug release which can impact on in vivo drug absorption. In vitro dissolution-permeation (D/P) methodologies that can account for the effects of enabling formulations on the permeability of drugs are increasingly being used in performance ranking during early development stages. This work comprised the application of two different cell-free in vitro D/P setups: BioFLUX™ and PermeaLoop™ to evaluate the dissolution-permeation interplay upon drug release from itraconazole (ITZ)- HPMCAS amorphous solid dispersions (ASDs) of different drug loads. A solvent-shift approach was employed, from a simulated gastric environment to a simulated intestinal environment in the donor compartment. PermeaLoop™ was then combined with microdialysis sampling to separate the dissolved (free) drug from other species present in solution, like micelle-bound drug and drug-rich colloids, in real time. This setup was applied to clarify the mechanisms for drug release and permeation from these ASDs. In parallel, a pharmacokinetic study (dog model) was conducted to assess the drug absorption from these ASDs and to compare the in vivo results with the data obtained from each in vitro D/P setup, allowing to infer which would be the most adequate setup for ASD ranking. Even though both D/P systems resulted in the same qualitative ranking, BioFLUX™ overpredicted the difference between the in vivo AUC of two ASDs, whereas PermeaLoop™ permeation flux resulted in a good correlation with the AUC observed in pharmacokinetic studies (dog model) (R2 ≈ 0.98). Also, PermeaLoop™ combined with a microdialysis sampling probe clarified the mechanisms for drug release and permeation from these ASDs. It demonstrated that the free drug was the only driving force for permeation, while the drug-rich colloids kept permeation active for longer periods by acting as drug reservoirs and maintaining constant high levels of free drug in solution, which are then immediately able to permeate. Hence, the data obtained points BioFLUX™ and PermeaLoop™ applications to different momentums in the drug product development pipeline: while BioFLUX™, an automated standardized method, poses as a valuable tool for initial ASD ranking during the early development stages, PermeaLoop™ combined with microdialysis sampling allows to gain mechanistic understanding of the dissolution-permeation interplay, being crucial to fine tune and identify leading ASD candidates prior to in vivo testing.
Collapse
Affiliation(s)
- Patrícia D Nunes
- R&D Analytical Development, Hovione Farmaciência S.A., Campus do Lumiar, Building S, 1649-038 Lisboa, Portugal; R&D Oral Drug Product Development, Hovione Farmaciência S.A., Campus do Lumiar, Building S, 1649-038 Lisboa, Portugal; Research Institute for Medicines (iMed.Ulisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Ana Filipa Ferreira
- R&D Analytical Development, Hovione Farmaciência S.A., Campus do Lumiar, Building S, 1649-038 Lisboa, Portugal
| | - João F Pinto
- Research Institute for Medicines (iMed.Ulisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal.
| | - Annette Bauer-Brandl
- Drug Transport and Delivery Group, Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense DK-5230, Denmark
| | - Martin Brandl
- Drug Transport and Delivery Group, Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense DK-5230, Denmark.
| | - João Henriques
- R&D Oral Drug Product Development, Hovione Farmaciência S.A., Campus do Lumiar, Building S, 1649-038 Lisboa, Portugal
| | - Ana Mafalda Paiva
- R&D Analytical Development, Hovione Farmaciência S.A., Campus do Lumiar, Building S, 1649-038 Lisboa, Portugal
| |
Collapse
|
49
|
Saha SK, Joshi A, Singh R, Dubey K. Review of industrially recognized polymers and manufacturing processes for amorphous solid dispersion based formulations. Pharm Dev Technol 2023; 28:678-696. [PMID: 37427544 DOI: 10.1080/10837450.2023.2233595] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/27/2023] [Accepted: 07/02/2023] [Indexed: 07/11/2023]
Abstract
Evolving therapeutic landscape through combinatorial chemistry and high throughput screening have resulted in an increased number of poorly soluble drugs. Drug delivery strategies quickly adapted to convert these drugs into successful therapies. Amorphous solid dispersion (ASD) technology is widely employed as a drug delivery strategy by pharmaceutical industries to overcome the challenges associated with these poorly soluble drugs. The development of ASD formulation requires an understanding of polymers and manufacturing techniques. A review of US FDA-approved ASD-based products revealed that only a limited number of polymers and manufacturing technologies are employed by pharmaceutical industries. This review provides a comprehensive guide for the selection and overview of polymers and manufacturing technologies adopted by pharmaceutical industries for ASD formulation. The various employed polymers with their underlying mechanisms for solution-state and solid-state stability are discussed. ASD manufacturing techniques, primarily implemented by pharmaceutical industries for commercialization, are presented in Quality by Design (QbD) format. An overview of novel excipients and progress in manufacturing technologies are also discussed. This review provides insights to the researchers on the industrially accepted polymers and manufacturing technology for ASD formulation that has translated these challenging drugs into successful therapies.
Collapse
Affiliation(s)
- Sumit Kumar Saha
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
- Formulation Research and Development - Orals, Sun Pharmaceuticals Industries Limited, Gurugram, India
| | | | - Romi Singh
- Formulation Research and Development - Orals, Sun Pharmaceuticals Industries Limited, Gurugram, India
| | - Kiran Dubey
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
50
|
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
|