1
|
Figueiredo J, Mendes M, Pais A, Sousa J, Vitorino C. Microfluidics-on-a-chip for designing celecoxib-based amorphous solid dispersions: when the process shapes the product. Drug Deliv Transl Res 2025; 15:732-752. [PMID: 38861140 PMCID: PMC11683022 DOI: 10.1007/s13346-024-01633-7] [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] [Accepted: 05/13/2024] [Indexed: 06/12/2024]
Abstract
The fundamental idea underlying the use of amorphous solid dispersions (ASDs) is to make the most of the solubility advantage of the amorphous form of a drug. However, the drug stability becomes compromised due to the higher free energy and disorder of molecular packing in the amorphous phase, leading to crystallization. Polymers are used as a matrix to form a stable homogeneous amorphous system to overcome the stability concern. The present work aims to design ASD-based formulations under the umbrella of quality by design principles for improving oral drug bioavailability, using celecoxib (CXB) as a model drug. ASDs were prepared from selected polymers and tested both individually and in combinations, using various manufacturing techniques: high-shear homogenization, high-pressure homogenization, microfluidics-on-a-chip, and spray drying. The resulting dispersions were further optimized, resorting to a 32 full-factorial design, considering the drug:polymers ratio and the total solid content as variables. The formulated products were evaluated regarding analytical centrifugation and the influence of the different polymers on the intrinsic dissolution rate of the CXB-ASDs. Microfluidics-on-a-chip led to the amorphous status of the formulation. The in vitro evaluation demonstrated a remarkable 26-fold enhancement in the intrinsic dissolution rate, and the translation of this formulation into tablets as the final dosage form is consistent with the observed performance enhancement. These findings are supported by ex vivo assays, which exhibited a two-fold increase in permeability compared to pure CXB. This study tackles the bioavailability hurdles encountered with diverse active compounds, offering insights into the development of more effective drug delivery platforms.
Collapse
Affiliation(s)
- Joana Figueiredo
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
| | - Maria Mendes
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, 3004-535, Coimbra, Portugal
| | - Alberto Pais
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, 3004-535, Coimbra, Portugal
| | - João Sousa
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, 3004-535, Coimbra, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal.
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, 3004-535, Coimbra, Portugal.
| |
Collapse
|
2
|
Hu H, Koranne S, Bower CM, Skomski D, Lamm MS. High-Speed Imaging-Based Particle Attribute Analysis of Spray-Dried Amorphous Solid Dispersions Using a Convolution Neural Network. Mol Pharm 2025; 22:488-497. [PMID: 39620431 DOI: 10.1021/acs.molpharmaceut.4c01092] [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: 01/07/2025]
Abstract
Spray drying is a well-established method for preparing amorphous solid dispersion (ASD) formulations to improve the oral bioavailability of poorly soluble drugs. In addition to the characterization of the amorphous phase, particle attributes of spray-dried intermediates (SDIs), including particle size, morphology, and microstructure, need to be carefully studied and controlled for optimizing drug product performance. Although recent developments in microscopy technology have enabled the analysis of morphological attributes for individual SDI particles, a high-throughput method is highly desirable. In this work, a fingerprinting method exploiting high-speed dynamic imaging, laser diffraction (LD), and a convolutional neural network (CNN) was developed to characterize and quantify size and morphological distributions of particles in batches of spray-dried ASDs. This imaging technology enables the generation of hundreds of thousands of single-particle images in a few minutes that are analyzed by both unsupervised and supervised CNN models. The unsupervised data mining analysis demonstrated that a batch of SDI is a mixture of diverse particle subpopulations with varying sizes and morphological attributes. Motivated by this observation, we developed a CNN model that enabled rapid computation of the volumetric composition of the distinct particle subpopulations in a SDI batch, thus generating a morphological fingerprint. We implemented this high-speed imaging-based particle attribute analysis method to investigate SDIs containing hypromellose acetate succinate as a model system. The CNN fingerprint results enabled quantification of the changes in the morphological distribution of SDI batches prepared with variations in the spray drying process parameters, and the results were in line with the LD and electron microscopy data. Our experiments and analysis demonstrate the robustness and throughput of this fingerprinting approach for quantifying particle size and morphological distributions of individual SDI batches, which can help guide spray drying process development and thereby enable the development of a drug product with more robust process and optimized performance.
Collapse
Affiliation(s)
- Hang Hu
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Sampada Koranne
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Colton M Bower
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Daniel Skomski
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Matthew S Lamm
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| |
Collapse
|
3
|
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
|
4
|
Su D, Bai M, Wei C, Long X, Liu X, Shen X, Ding H. Combining solubilization and controlled release strategies to prepare pH-sensitive solid dispersion loaded with albendazole: in vitro and in vivo studies. Front Vet Sci 2024; 11:1522856. [PMID: 39758610 PMCID: PMC11695277 DOI: 10.3389/fvets.2024.1522856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/11/2024] [Indexed: 01/07/2025] Open
Abstract
Albendazole (ABZ), classified as a class II basic drug under the Biopharmaceutics Classification System (BCS), is widely recognized for its therapeutic efficacy in treating and preventing trichuriasis. However, despite its clinical relevance, ABZ's oral administration presents challenges due to its poor solubility and pH sensitivity, which diminish its therapeutic effectiveness. Additionally, high dosing regimens of ABZ pose risks of developmental toxicity in animal models. This study developed a pH-sensitive solid dispersion of albendazole (ABZ-pHs-SD) using Glyceryl Monostearate (GM) in conjunction with Hypromellose Acetate Succinate (HPMC-AS). Characterization via Scanning Electron Microscopy (SEM), Powder X-ray Diffraction (PXRD), Differential Scanning Calorimetry (DSC), and Fourier Transform Infrared Spectroscopy (FT-IR) confirmed the high dispersion of ABZ in a crystalline state within the carrier. Furthermore, we compared the in vitro dissolution profile, pharmacokinetics, and intestinal drug concentration of ABZ-pHs-SD with commercially available formulations. Our findings demonstrated that ABZ-pHs-SD exhibited an excellent dissolution profile, significantly increasing the solubility of ABZ in water by 3.15 times. The formulation effectively prevented drug release in acidic environments while maintaining a slow release in weakly alkaline conditions. Additionally, compared to commercial formulations, ABZ-pHs-SD showed significantly lower Cmax (4.70 ± 1.16 vs. 6.83 ± 0.66 μg/mL) and higher Tmax (5.5 ± 0.93 vs. 3.75 ± 0.71 h) in vivo, achieving elevated drug concentration levels in the cecal and colonic environments (p < 0.01) without significantly decreasing bioavailability. Overall, our research findings indicate that ABZ-pHs-SD serves as a promising drug delivery strategy for the poorly soluble and pH-sensitive ABZ. Particularly, the simple preparation of solid dispersion demonstrates strong industrial feasibility.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Huanzhong Ding
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University (SCAU), Guangzhou, China
| |
Collapse
|
5
|
Khorami K, Farahani SD, Müllertz A, Rades T. Drug-Phospholipid Co-Amorphous Formulations: The Role of Preparation Methods and Phospholipid Selection. Pharmaceutics 2024; 16:1602. [PMID: 39771580 PMCID: PMC11676457 DOI: 10.3390/pharmaceutics16121602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: This study aims to broaden the knowledge on co-amorphous phospholipid systems (CAPSs) by exploring the formation of CAPSs with a broader range of poorly water-soluble drugs, celecoxib (CCX), furosemide (FUR), nilotinib (NIL), and ritonavir (RIT), combined with amphiphilic phospholipids (PLs), including soybean phosphatidylcholine (SPC), hydrogenated phosphatidylcholine (HPC), and mono-acyl phosphatidylcholine (MAPC). Methods: The CAPSs were initially prepared at equimolar drug-to-phospholipid (PL) ratios by mechano-chemical activation-based, melt-based, and solvent-based preparation methods, i.e., ball milling (BM), quench cooling (QC), and solvent evaporation (SE), respectively. The solid state of the product was characterized by X-ray powder diffraction (XRPD), polarized light microscopy (PLM), and differential scanning calorimetry (DSC). The long-term physical stability of the CAPSs was investigated at room temperature under dry conditions (0% RH) and at 75% RH. The dissolution behavior of the CCX CAPS and RIT CAPS was studied. Results: Our findings indicate that SE consistently prepared CAPSs for CCX-PLs, FUR-PLs, and RIT-PLs, whereas the QC method could only form CAPSs for RIT-PLs, CCX-SPC, and CCX-MAPC. In contrast, the BM method failed to produce CAPSs, but all drugs alone could be fully amorphized. While the stability of each drug varied depending on the PLs used, the SE CAPS consistently demonstrated the highest stability by a significant margin. Initially, a 1:1 molar ratio was used for screening all systems, though the optimal molar ratio for drug stability remained uncertain. To address this, various molar ratios were investigated to determine the ratio yielding the highest amorphous drug stability. Our results indicate that all systems remained physically stable at a 1.5:1 ratio and with excess of PL. Furthermore, the CAPS formed by the SE significantly improves the dissolution behavior of CCX and RIT, whereas the PLs provide a slight precipitation inhibition for supersaturated CCX and RIT. Conclusions: These findings support the use of a 1:1 molar ratio in screening processes and suggest that CAPSs can be effectively prepared with relatively high drug loads compared to traditional drug-polymer systems. Furthermore, the study highlights the critical role of drug selection, the preparation method, and the PL type in developing stable and effective CAPSs.
Collapse
Affiliation(s)
| | | | - Anette Müllertz
- Department of Pharmacy, Faculty of Health and Medical Science, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; (K.K.); (S.D.F.); (T.R.)
| | | |
Collapse
|
6
|
Parkes A, Ziaee A, O'Reilly E. Evaluating experimental, knowledge-based and computational cocrystal screening methods to advance drug-drug cocrystal fixed-dose combination development. Eur J Pharm Sci 2024; 203:106931. [PMID: 39389169 DOI: 10.1016/j.ejps.2024.106931] [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: 07/15/2024] [Revised: 09/18/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
Fixed-dose combinations (FDCs) offer significant advantages to patients and the pharmaceutical industry alike through improved dissolution profiles, synergistic effects and extended patent lifetimes. Identifying whether two active pharmaceutical ingredients have the potential to form a drug-drug cocrystal (DDC) or interact is an essential step in determining the most suitable type of FDC to formulate. The lack of coherent strategies to determine if two active pharmaceutical ingredients that can be co-administered can form a cocrystal, has significantly impacted DDC commercialisation. This review aims to accelerate the development of FDCs and DDCs by evaluating existing experimental, knowledge-based and computational cocrystal screening methods; the background of their development, their application in screening for cocrystals and DDCs, and their limitations are discussed. The evaluation provided in this review will act as a guide for selecting suitable screening methods to accelerate FDC development.
Collapse
Affiliation(s)
- Alice Parkes
- Department of Chemical Sciences, SSPC the SFI Research Centre for Pharmaceuticals, Bernal Institute, University of Limerick, Limerick, Ireland
| | | | - Emmet O'Reilly
- Department of Chemical Sciences, SSPC the SFI Research Centre for Pharmaceuticals, Bernal Institute, University of Limerick, Limerick, Ireland.
| |
Collapse
|
7
|
Kim HJ, Jang J, Lee J, Han CH, Kim JW, Park BJ. Fabrication of Engineered Drug-Polymer Composite Particles via Piezoelectric Inkjet Technique for Floating Drug Delivery Systems. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024. [PMID: 39556093 DOI: 10.1021/acs.langmuir.4c03556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
This study presents a simple approach for fabricating low-density drug-polymer amorphous solid dispersions (ASDs) using a piezoelectric inkjet method, demonstrating potential applications for floating drug delivery systems (FDDS). By adjusting the ratio of two polymers, polylactic acid, and Eudragit RLPO, the floatability and drug release rate of the drug-polymer ASD particles can be easily manipulated. Kinetic model analyses have been conducted to interpret the drug release mechanism. This work offers a robust platform for exploring diverse polymer-drug combinations that are applicable to FDDS.
Collapse
Affiliation(s)
- Hee Jin Kim
- Department of Chemical Engineering (BK21 FOUR Integrated Engineering Program), Kyung Hee University, Yongin 17104, South Korea
| | - Jiye Jang
- Department of Chemical Engineering (BK21 FOUR Integrated Engineering Program), Kyung Hee University, Yongin 17104, South Korea
| | - Jieun Lee
- Department of Chemical Engineering (BK21 FOUR Integrated Engineering Program), Kyung Hee University, Yongin 17104, South Korea
| | - Chang Hun Han
- Department of Chemical Engineering (BK21 FOUR Integrated Engineering Program), Kyung Hee University, Yongin 17104, South Korea
| | - Jin Woong Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Bum Jun Park
- Department of Chemical Engineering (BK21 FOUR Integrated Engineering Program), Kyung Hee University, Yongin 17104, South Korea
| |
Collapse
|
8
|
Nyamba I, Sombié CB, Yabré M, Zimé-Diawara H, Yaméogo J, Ouédraogo S, Lechanteur A, Semdé R, Evrard B. Pharmaceutical approaches for enhancing solubility and oral bioavailability of poorly soluble drugs. Eur J Pharm Biopharm 2024; 204:114513. [PMID: 39313163 DOI: 10.1016/j.ejpb.2024.114513] [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: 06/23/2024] [Revised: 09/15/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024]
Abstract
High solubility in water and physiological fluids is an indispensable requirement for the pharmacological efficacy of an active pharmaceutical ingredient. Indeed, it is well established that pharmaceutical substances exhibiting limited solubility in water are inclined towards diminished and inconsistent absorption following oral administration, consequently resulting in variability in therapeutic outcomes. The current advancements in combinatorial chemistry and pharmaceutical design have facilitated the creation of drug candidates characterized by increased lipophilicity, elevated molecular size, and reduced aqueous solubility. Undoubtedly, the issue of poorly water-soluble medications has been progressively escalating over recent years. Indeed, 40% of the top 200 oral medications marketed in the United States, 33% of drugs listed in the US pharmacopoeia, 75% of compounds under development and 90% of new chemical entities are insufficiently water-soluble compounds. In order to address this obstacle, formulation scientists employ a variety of approaches, encompassing both physical and chemical methods such as prodrug synthesis, salt formation, solid dispersions formation, hydrotropic substances utilization, solubilizing agents incorporation, cosolvent addition, polymorphism exploration, cocrystal creation, cyclodextrins complexation, lipid formulations, particle size reduction and nanoformulation techniques. Despite the utilization of these diverse approaches, the primary reason for the failure in new drug development persists as the poor aqueous solubility of pharmaceutical compounds. This paper, therefore, delves into the foundational principles that underpin the implementation of various formulation strategies, along with a discussion on the respective advantages and drawbacks associated with each approach. Additionally, a discourse is provided regarding methodological frameworks for making informed decisions on selecting an appropriate formulation strategy to effectively tackle the key challenges posed during the development of a poorly water-soluble drug candidate.
Collapse
Affiliation(s)
- Isaïe Nyamba
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, Université de Liège, 4000 Liège, Belgium; Laboratory of Drug Development, Center of Training, Research and Expertise in Pharmaceutical Sciences (CFOREM), Doctoral School of Sciences and Health, Université Joseph KI-ZERBO, 03 BP 7021 Ouagadougou 03, Burkina Faso; Institut Supérieur des Sciences de la Santé (INSSA), Université Nazi Boni, 01 BP 1091 Bobo-Dioulasso 01, Burkina Faso.
| | - Charles B Sombié
- Laboratory of Drug Development, Center of Training, Research and Expertise in Pharmaceutical Sciences (CFOREM), Doctoral School of Sciences and Health, Université Joseph KI-ZERBO, 03 BP 7021 Ouagadougou 03, Burkina Faso
| | - Moussa Yabré
- Institut Supérieur des Sciences de la Santé (INSSA), Université Nazi Boni, 01 BP 1091 Bobo-Dioulasso 01, Burkina Faso
| | - Hermine Zimé-Diawara
- Laboratory of Drug Development, Center of Training, Research and Expertise in Pharmaceutical Sciences (CFOREM), Doctoral School of Sciences and Health, Université Joseph KI-ZERBO, 03 BP 7021 Ouagadougou 03, Burkina Faso
| | - Josias Yaméogo
- Laboratory of Drug Development, Center of Training, Research and Expertise in Pharmaceutical Sciences (CFOREM), Doctoral School of Sciences and Health, Université Joseph KI-ZERBO, 03 BP 7021 Ouagadougou 03, Burkina Faso
| | - Salfo Ouédraogo
- Laboratory of Drug Development, Center of Training, Research and Expertise in Pharmaceutical Sciences (CFOREM), Doctoral School of Sciences and Health, Université Joseph KI-ZERBO, 03 BP 7021 Ouagadougou 03, Burkina Faso
| | - Anna Lechanteur
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, Université de Liège, 4000 Liège, Belgium
| | - Rasmané Semdé
- Laboratory of Drug Development, Center of Training, Research and Expertise in Pharmaceutical Sciences (CFOREM), Doctoral School of Sciences and Health, Université Joseph KI-ZERBO, 03 BP 7021 Ouagadougou 03, Burkina Faso
| | - Brigitte Evrard
- Laboratory of Pharmaceutical Technology and Biopharmacy, CIRM, Université de Liège, 4000 Liège, Belgium
| |
Collapse
|
9
|
Alvebratt C, Karlén F, Åhlén M, Edueng K, Dubbelboer I, Bergström CAS. Benefits of combining supersaturating and solubilizing formulations - Is two better than one? Int J Pharm 2024; 663:124437. [PMID: 39002818 DOI: 10.1016/j.ijpharm.2024.124437] [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: 05/07/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/15/2024]
Abstract
A variety of enabling formulations has been developed to address poor oral drug absorption caused by insufficient dissolution in the gastrointestinal tract. As the in vivo performance of these formulations is a result of a complex interplay between dissolution, digestion and permeation, development of suitable in vitro assays that captures these phenomena are called for. The enabling-absorption (ENA) device, consisting of a donor and receiver chamber separated by a semipermeable membrane, has successfully been used to study the performance of lipid-based formulations. In this work, the ENA device was prepared with two different setups (a Caco-2 cell monolayer and an artificial lipid membrane) to study the performance of a lipid-based formulation (LBF), an amorphous solid dispersion (ASD) and the potential benefit of combining the two formulation strategies. An in vivo pharmacokinetic study in rats was performed to evaluate the in vitro-in vivo correlation. In the ENA, high drug concentrations in the donor chamber did not translate to a high mass transfer, which was particularly evident for the ASD as compared to the LBF. The solubility of the polymer used in the ASD was strongly affected by pH-shifts in vitro, and the ph_dependence resulted in poor in vivo performance of the formulation. The dissolution was however increased in vitro when the ASD was combined with a blank lipid-based formulation. This beneficial effect was also observed in vivo, where the drug exposure of the ASD increased significantly when the ASD was co-administered with the blank LBF. To conclude, the in vitro model managed to capture solubility limitations and strategies to overcome these for one of the formulations studied. The correlation between the in vivo exposure of the drug exposure and AUC in the ENA was good for the non pH-sensitive formulations. The deconvoluted pharmacokinetic data indicated that the receiver chamber was a better predictor for the in vivo performance of the drug, however both chambers provided valuable insights to the observed outcome in vivo. This shows that the advanced in vitro setting used herein successfully could explain absorption differences of highly complex formulations.
Collapse
Affiliation(s)
- Caroline Alvebratt
- Department of Pharmacy, Uppsala Biomedical Centre, P.O. Box 580, Uppsala University, Uppsala SE-751 23, Sweden.
| | - Filip Karlén
- Department of Pharmacy, Uppsala Biomedical Centre, P.O. Box 580, Uppsala University, Uppsala SE-751 23, Sweden.
| | - Michelle Åhlén
- Division of Nanotechnology and Functional Materials, Department of Engineering Sciences, Uppsala University, Uppsala SE-75121, Sweden.
| | - Khadijah Edueng
- Department of Pharmacy, Uppsala Biomedical Centre, P.O. Box 580, Uppsala University, Uppsala SE-751 23, Sweden
| | - Ilse Dubbelboer
- The Swedish Drug Delivery Center, Department of Pharmaceutical Biosciences, Uppsala Biomedical Centre, P.O. Box 591, Uppsala University, Uppsala SE-751 24, Sweden.
| | - Christel A S Bergström
- Department of Pharmacy, Uppsala Biomedical Centre, P.O. Box 580, Uppsala University, Uppsala SE-751 23, Sweden; The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala Biomedical Centre, P.O. Box 580, Uppsala University, Uppsala SE-751 23, Sweden.
| |
Collapse
|
10
|
Di Mare EJ, Punia A, Lamm MS, Rhodes TA, Gormley AJ. Data-Driven Design of Novel Polymer Excipients for Pharmaceutical Amorphous Solid Dispersions. Bioconjug Chem 2024; 35:1363-1372. [PMID: 39150455 DOI: 10.1021/acs.bioconjchem.4c00294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
About 90% of active pharmaceutical ingredients (APIs) in the oral drug delivery system pipeline have poor aqueous solubility and low bioavailability. To address this problem, amorphous solid dispersions (ASDs) embed hydrophobic APIs within polymer excipients to prevent drug crystallization, improve solubility, and increase bioavailability. There are a limited number of commercial polymer excipients, and the structure-function relationships which lead to successful ASD formulations are not well-documented. There are, however, certain solid-state ASD characteristics that inform ASD performance. One characteristic shared by successful ASDs is a high glass transition temperature (Tg), which correlates with higher shelf stability and decreased drug crystallization. We aim to identify how polymer features such as side chain geometry, backbone methylation, and hydrophilic-lipophilic balance impact Tg to design copolymers capable of forming high-Tg ASDs. We tested a library of 50 ASD formulations (18 previously studied and 32 newly synthesized) of the model drug probucol with copolymers synthesized through automated photoinduced electron/energy transfer-reversible addition-fragmentation chain-transfer (PET-RAFT) polymerization. A machine learning (ML) algorithm was trained on the Tg data to identify the major factors influencing Tg, including backbone methylation and nonlinear side chain geometry. In both polymer alone and probucol-loaded ASDs, a Random Forest Regressor captured structure-function trends in the data set and accurately predicted Tg with an average R2 > 0.83 across a 10-fold cross validation. This ML model will be used to predict novel copolymers to design ASDs with high Tg, a crucial factor in predicting ASD success.
Collapse
Affiliation(s)
- Elena J Di Mare
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Ashish Punia
- Analytical Research and Development, MRL, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Matthew S Lamm
- Analytical Research and Development, MRL, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Timothy A Rhodes
- Analytical Research and Development, MRL, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Adam J Gormley
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| |
Collapse
|
11
|
Gilbert E, Salonen A, Poulard C. Decoupling the rheological responses of a soft solid emulsion with liquid inclusions. JOURNAL OF PHYSICS. CONDENSED MATTER : AN INSTITUTE OF PHYSICS JOURNAL 2024; 36:425103. [PMID: 38986471 DOI: 10.1088/1361-648x/ad61ad] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/10/2024] [Indexed: 07/12/2024]
Abstract
Soft solid emulsions are liquid droplets encapsulated in a soft solid material. Typical of dispersed systems, they can combine properties from both the liquid inclusions and the soft solids. The relative importance of the two phases in the rheological response is captured through the elastocapillary number, which compares capillary forces in the liquid inclusions to the matrix rigidity. We work with solid emulsions formed of poly(ethylene glycol) droplets in a poly(dimethylsiloxane) (PDMS) continuous phase. We create three families of emulsions with varying elastocapillary numbers, and range of inclusion volume fractions from 0 to 0.5. Through oscillatory rheology we probe both the elastic response and the dissipative effects of liquid droplets. In the case of a dominant response from the continuous phase or the drops, the results can be described with Palierne's model. However, for the intermediate elastocapillary series we show that the evolution of the storage and loss moduli decouple with dispersed phase volume fraction. We attribute the increase of loss factor with volume fraction to the high polydispersity in droplet size. We can further modulate the response of the materials by cooling to freeze the droplets. This approach allows us to compare these soft solid emulsions with theories related to solid dispersions.
Collapse
Affiliation(s)
- E Gilbert
- INRAE, AgroParisTech, UMR SayFood, Université Paris-Saclay, Palaiseau 91120, France
- Université Paris-Saclay, CNRS, Laboratoire de Physique des Solides, Orsay 91400, France
| | - A Salonen
- Université Paris-Saclay, CNRS, Laboratoire de Physique des Solides, Orsay 91400, France
| | - C Poulard
- Université Paris-Saclay, CNRS, Laboratoire de Physique des Solides, Orsay 91400, France
| |
Collapse
|
12
|
Helmy AM, Lu A, Duggal I, Rodrigues KP, Maniruzzaman M. Electromagnetic drop-on-demand (DoD) technology as an innovative platform for amorphous solid dispersion production. Int J Pharm 2024; 658:124185. [PMID: 38703932 DOI: 10.1016/j.ijpharm.2024.124185] [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: 01/12/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/06/2024]
Abstract
Production of amorphous solid dispersions (ASDs) is an effective strategy to promote the solubility and bioavailability of poorly water soluble medicinal substances. In general, ASD is manufactured using a variety of classic and modern techniques, most of which rely on either melting or solvent evaporation. This proof-of-concept study is the first ever to introduce electromagnetic drop-on-demand (DoD) technique as an alternative solvent evaporation-based method for producing ASDs. Herein 3D printing of ASDs for three drug-polymer combinations (efavirenz-Eudragit L100-55, lumefantrine-hydroxypropyl methylcellulose acetate succinate, and favipiravir-polyacrylic acid) was investigated to ascertain the reliability of this technique. Polarized light microscopy, differential scanning calorimetry (DSC), X-ray powder diffraction (XRPD), and Fourier Transform Infrared (FTIR) spectroscopy results supported the formation of ASDs for the three drugs by means of DoD 3D printing, which significantly increases the equilibrium solubility of efavirenz from 0.03 ± 0.04 µg/ml to 21.18 ± 4.20 µg/ml, and the equilibrium solubility of lumefantrine from 1.26 ± 1.60 µg/ml to 20.21 ± 6.91 µg/ml. Overall, the reported findings show how this new electromagnetic DoD technology can have a potential to become a cutting-edge 3D printing solvent-evaporation technique for on-demand and continuous manufacturing of ASDs for a variety of drugs.
Collapse
Affiliation(s)
- Abdelrahman M Helmy
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Deraya University, Minya, Egypt
| | - Anqi Lu
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Ishaan Duggal
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Kristina P Rodrigues
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Mohammed Maniruzzaman
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38677-1848, USA.
| |
Collapse
|
13
|
Wang H, Zhao P, Ma R, Jia J, Fu Q. Drug-drug co-amorphous systems: An emerging formulation strategy for poorly water-soluble drugs. Drug Discov Today 2024; 29:103883. [PMID: 38219970 DOI: 10.1016/j.drudis.2024.103883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/21/2023] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
Overcoming the poor water solubility of small-molecule drugs is a major challenge in the development of clinical pharmaceuticals. Amorphization of crystalline drugs is a highly effective strategy to improve their aqueous solubility. However, amorphous drugs are thermodynamically unstable and likely to crystallize during manufacturing and storage. Recently, drug-drug co-amorphous systems have emerged as a novel strategy to not only enable enhanced dissolution and physical stability of the individual drugs within the system but also to provide a strategy for combination therapy of the same or different clinical indications. This review serves to highlight advances in the methods used to manufacture and characterize drug-drug co-amorphous systems, summarize drug-drug co-amorphous applications reported in recent decades, and provide an outlook on future possibilities and perspectives.
Collapse
Affiliation(s)
- Hongge Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Peixu Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Ruilong Ma
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Jirun Jia
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Qiang Fu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
14
|
Rusdin A, Mohd Gazzali A, Ain Thomas N, Megantara S, Aulifa DL, Budiman A, Muchtaridi M. Advancing Drug Delivery Paradigms: Polyvinyl Pyrolidone (PVP)-Based Amorphous Solid Dispersion for Enhanced Physicochemical Properties and Therapeutic Efficacy. Polymers (Basel) 2024; 16:286. [PMID: 38276694 PMCID: PMC10820039 DOI: 10.3390/polym16020286] [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: 11/28/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND The current challenge in drug development lies in addressing the physicochemical issues that lead to low drug effectiveness. Solubility, a crucial physicochemical parameter, greatly influences various biopharmaceutical aspects of a drug, including dissolution rate, absorption, and bioavailability. Amorphous solid dispersion (ASD) has emerged as a widely explored approach to enhance drug solubility. OBJECTIVE The objective of this review is to discuss and summarize the development of polyvinylpyrrolidone (PVP)-based amorphous solid dispersion in improving the physicochemical properties of drugs, with a focus on the use of PVP as a novel approach. METHODOLOGY This review was conducted by examining relevant journals obtained from databases such as Scopus, PubMed, and Google Scholar, since 2018. The inclusion and exclusion criteria were applied to select suitable articles. RESULTS This study demonstrated the versatility and efficacy of PVP in enhancing the solubility and bioavailability of poorly soluble drugs. Diverse preparation methods, including solvent evaporation, melt quenching, electrospinning, coprecipitation, and ball milling are discussed for the production of ASDs with tailored characteristics. CONCLUSION PVP-based ASDs could offer significant advantages in the formulation strategies, stability, and performance of poorly soluble drugs to enhance their overall bioavailability. The diverse methodologies and findings presented in this review will pave the way for further advancements in the development of effective and tailored amorphous solid dispersions.
Collapse
Affiliation(s)
- Agus Rusdin
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjadjaran, Jl. Raya Bandung-Sumedang Km-21, Bandung 45363, Indonesia; (A.R.); (S.M.); (D.L.A.)
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjadjaran, Jl. Raya Bandung-Sumedang Km-21, Bandung 45363, Indonesia;
| | - Amirah Mohd Gazzali
- Departement Pharmaceutical Technology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, P.Penang, Penang 11800, Malaysia;
| | - Nur Ain Thomas
- Department of Pharmacy, Faculty of Sport and Health, Universitas Negeri Gorontalo, Jl. Jenderal Sudirman No. 6, Gorontalo 96128, Indonesia;
| | - Sandra Megantara
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjadjaran, Jl. Raya Bandung-Sumedang Km-21, Bandung 45363, Indonesia; (A.R.); (S.M.); (D.L.A.)
- Research Collaboration Centre for Theranostic Radiopharmaceuticals, National Research and Innovation Agency (BRIN), Jakarta Pusat 10340, Indonesia
| | - Diah Lia Aulifa
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjadjaran, Jl. Raya Bandung-Sumedang Km-21, Bandung 45363, Indonesia; (A.R.); (S.M.); (D.L.A.)
| | - Arif Budiman
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjadjaran, Jl. Raya Bandung-Sumedang Km-21, Bandung 45363, Indonesia;
| | - Muchtaridi Muchtaridi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjadjaran, Jl. Raya Bandung-Sumedang Km-21, Bandung 45363, Indonesia; (A.R.); (S.M.); (D.L.A.)
- Research Collaboration Centre for Theranostic Radiopharmaceuticals, National Research and Innovation Agency (BRIN), Jakarta Pusat 10340, Indonesia
| |
Collapse
|
15
|
Tripathi D, B H MP, Sahoo J, Kumari J. Navigating the Solution to Drug Formulation Problems at Research and Development Stages by Amorphous Solid Dispersion Technology. RECENT ADVANCES IN DRUG DELIVERY AND FORMULATION 2024; 18:79-99. [PMID: 38062659 DOI: 10.2174/0126673878271641231201065151] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/20/2023] [Accepted: 10/26/2023] [Indexed: 08/30/2024]
Abstract
Amorphous Solid Dispersions (ASDs) have indeed revolutionized the pharmaceutical industry, particularly in drug solubility enhancement. The amorphous state of a drug, which is a highenergy metastable state, can lead to an increase in the apparent solubility of the drug. This is due to the absence of a long-range molecular order, which results in higher molecular mobility and free volume, and consequently, higher solubility. The success of ASD preparation depends on the selection of appropriate excipients, particularly polymers that play a crucial role in drug solubility and physical stability. However, ASDs face challenges due to their thermodynamic instability or tendency to recrystallize. Measuring the crystallinity of the active pharmaceutical ingredient (API) and drug solubility is a complex process that requires a thorough understanding of drug-polymer miscibility and molecular interactions. Therefore, it is important to monitor drug solids closely during preparation, storage, and application. Techniques such as solid-state nuclear magnetic resonance (ssNMR), attenuated total reflectance Fourier transform infrared spectroscopy (ATR-FTIR), Raman spectroscopy, and dielectric spectroscopy have been successful in understanding the mechanism of drug crystallization. In addition, the continuous downstream processing of drug-loaded ASDs has introduced new automated methods for consistent ASD production. Advanced techniques such as hot melt extrusion, KinetiSol, electro spraying, and electrospinning have gained popularity. This review provides a comprehensive overview of Amorphous Solid Dispersions (ASDs) for oral drug delivery. It highlights the critical challenges faced during formulation, the impact of manufacturing variables, theoretical aspects of drug-polymer interaction, and factors related to drug-polymer miscibility. ASDs have been recognized as a promising strategy to improve the oral bioavailability of poorly water-soluble drugs. However, the successful development of an ASD-based drug product is not straightforward due to the complexity of the ASD systems. The formulation and process parameters can significantly influence the performance of the final product. Understanding the interactions between the drug and polymer in ASDs is crucial for predicting their stability and performance.
Collapse
Affiliation(s)
- Devika Tripathi
- Pranveer Singh Institute of Technology (Pharmacy), Uttar Pradesh, Kanpur, India
| | - Manjunatha Prabhu B H
- Department of Food Protection and Infestation Control, CSIR-CFTRI, Central Food Technological Research Institute, Mysore, India
| | - Jagannath Sahoo
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, NIMMS, Mumbai, India
| | - Jyoti Kumari
- Pranveer Singh Institute of Technology (Pharmacy), Uttar Pradesh, Kanpur, India
| |
Collapse
|
16
|
Rantanen J, Rades T, Strachan C. Solid-state analysis for pharmaceuticals: Pathways to feasible and meaningful analysis. J Pharm Biomed Anal 2023; 236:115649. [PMID: 37657177 DOI: 10.1016/j.jpba.2023.115649] [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: 05/18/2023] [Revised: 08/12/2023] [Accepted: 08/13/2023] [Indexed: 09/03/2023]
Abstract
The solid state of matter is the preferred starting point for designing a pharmaceutical product. This is driven by both patient preferences and the relative ease of supplying a solid pharmaceutical product with desired quality and performance. Solid form diversity is increasingly prevalent as a crucial element in designing these products, which underpins the importance of solid-state analytical methods. This paper provides a critical analysis of challenges related to solid-state analytics, as well as considerations and suggestions for feasible and meaningful pharmaceutical analysis.
Collapse
Affiliation(s)
- Jukka Rantanen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| | - Thomas Rades
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| | | |
Collapse
|
17
|
Hatipoglu MK, Zaker Y, Willett DR, Gupta N, Rodriguez JD, Patankar S, Capella P, Yilmaz H. Old Polymorph, New Technique: Assessing Ritonavir Crystallinity Using Low-Frequency Raman Spectroscopy. Anal Chem 2023; 95:15325-15332. [PMID: 37796650 DOI: 10.1021/acs.analchem.3c02781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Two decades ago, postmarket discovery of a second crystal form of ritonavir with lower solubility had major implications for drug manufacturers and patients. Since then, ritonavir has been reformulated via the hot-melt-extrusion process in an amorphous form. Here, quantitative low- and mid-frequency Raman spectroscopy methods were developed to characterize polymorphs, form I and form II, in commercial ritonavir 100 mg oral tablets as an alternate analysis approach compared to X-ray powder diffraction (XRPD). Crystallization in three lots of ritonavir products obtained from four separate manufacturers was assessed after storage under accelerated conditions at 40 °C and 75% relative humidity (RH). Results were compared with quantitative XRPD methods developed and validated according to ICH Q2 (R1) guidelines. In a four-week open-dish study, form I crystallization occurred in two of the four products and form II crystallization was detected in another ritonavir product. The limits of detection for XRPD, low-frequency Raman (LFR), and mid-frequency Raman (MFR) were determined to be 0.7, 0.8, and 0.5% for form I and 0.6, 0.6, and 1% for form II, respectively. Root-mean-squared-error of predictions were 0.6-1.0 and 0.6-2.5% for LFR- and MFR-based partial least-squares models. Further, ritonavir polymorphs could also be identified and detected directly from ritonavir tablets using transmission LFR. In summary, LFR was applied for the assessment of polymorphism in real-world samples. While providing analytical performance similar to conventional techniques, LFR reduced the single measurement time from 66 min (XRPD) to 10 s (LFR) without the need for tedious sample preparation procedures.
Collapse
Affiliation(s)
- Manolya K Hatipoglu
- Division of Complex Drug Analysis, Office of Testing and Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, St. Louis, Missouri 63110, United States
| | - Yeakub Zaker
- Division of Complex Drug Analysis, Office of Testing and Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, St. Louis, Missouri 63110, United States
| | - Daniel R Willett
- Division of Complex Drug Analysis, Office of Testing and Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, St. Louis, Missouri 63110, United States
| | - Nirzari Gupta
- Division of Complex Drug Analysis, Office of Testing and Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, St. Louis, Missouri 63110, United States
| | - Jason D Rodriguez
- Division of Complex Drug Analysis, Office of Testing and Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, St. Louis, Missouri 63110, United States
| | - Suhas Patankar
- Division of Immediate & Modified Release Products II, Office of Lifecycle Drug Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Peter Capella
- Division of Immediate & Modified Release Products II, Office of Lifecycle Drug Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Huzeyfe Yilmaz
- Division of Complex Drug Analysis, Office of Testing and Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, St. Louis, Missouri 63110, United States
| |
Collapse
|
18
|
Budiman A, Lailasari E, Nurani NV, Yunita EN, Anastasya G, Aulia RN, Lestari IN, Subra L, Aulifa DL. Ternary Solid Dispersions: A Review of the Preparation, Characterization, Mechanism of Drug Release, and Physical Stability. Pharmaceutics 2023; 15:2116. [PMID: 37631330 PMCID: PMC10459848 DOI: 10.3390/pharmaceutics15082116] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
The prevalence of active pharmaceutical ingredients (APIs) with low water solubility has experienced a significant increase in recent years. These APIs present challenges in formulation, particularly for oral dosage forms, despite their considerable therapeutic potential. Therefore, the improvement of solubility has become a major concern for pharmaceutical enterprises to increase the bioavailability of APIs. A promising formulation approach that can effectively improve the dissolution profile and the bioavailability of poorly water-soluble drugs is the utilization of amorphous systems. Numerous formulation methods have been developed to enhance poorly water-soluble drugs through amorphization systems, including co-amorphous formulations, amorphous solid dispersions (ASDs), and the use of mesoporous silica as a carrier. Furthermore, the successful enhancement of certain drugs with poor aqueous solubility through amorphization has led to their incorporation into various commercially available preparations, such as ASDs, where the crystalline structure of APIs is transformed into an amorphous state within a hydrophilic matrix. A novel approach, known as ternary solid dispersions (TSDs), has emerged to address the solubility and bioavailability challenges associated with amorphous drugs. Meanwhile, the introduction of a third component in the ASD and co-amorphous systems has demonstrated the potential to improve performance in terms of solubility, physical stability, and processability. This comprehensive review discusses the preparation and characterization of poorly water-soluble drugs in ternary solid dispersions and their mechanisms of drug release and physical stability.
Collapse
Affiliation(s)
- Arif Budiman
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (E.L.); (N.V.N.); (E.N.Y.); (G.A.); (R.N.A.)
| | - Eli Lailasari
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (E.L.); (N.V.N.); (E.N.Y.); (G.A.); (R.N.A.)
| | - Neng Vera Nurani
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (E.L.); (N.V.N.); (E.N.Y.); (G.A.); (R.N.A.)
| | - Ellen Nathania Yunita
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (E.L.); (N.V.N.); (E.N.Y.); (G.A.); (R.N.A.)
| | - Gracia Anastasya
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (E.L.); (N.V.N.); (E.N.Y.); (G.A.); (R.N.A.)
| | - Rizqa Nurul Aulia
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (E.L.); (N.V.N.); (E.N.Y.); (G.A.); (R.N.A.)
| | - Ira Novianty Lestari
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (I.N.L.); (D.L.A.)
| | - Laila Subra
- Faculty of Bioeconomic and Health Sciences, Geomatika University College, Kuala Lumpur 54200, Malaysia;
| | - Diah Lia Aulifa
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (I.N.L.); (D.L.A.)
| |
Collapse
|
19
|
Zhang J, Guo M, Luo M, Cai T. Advances in the development of amorphous solid dispersions: The role of polymeric carriers. Asian J Pharm Sci 2023; 18:100834. [PMID: 37635801 PMCID: PMC10450425 DOI: 10.1016/j.ajps.2023.100834] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/26/2023] [Accepted: 07/23/2023] [Indexed: 08/27/2023] Open
Abstract
Amorphous solid dispersion (ASD) is one of the most effective approaches for delivering poorly soluble drugs. In ASDs, polymeric materials serve as the carriers in which the drugs are dispersed at the molecular level. To prepare the solid dispersions, there are many polymers with various physicochemical and thermochemical characteristics available for use in ASD formulations. Polymer selection is of great importance because it influences the stability, solubility and dissolution rates, manufacturing process, and bioavailability of the ASD. This review article provides a comprehensive overview of ASDs from the perspectives of physicochemical characteristics of polymers, formulation designs and preparation methods. Furthermore, considerations of safety and regulatory requirements along with the studies recommended for characterizing and evaluating polymeric carriers are briefly discussed.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China
| | - Minshan Guo
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Minqian Luo
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Ting Cai
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
20
|
Suhaidi D, Dong YD, Wynne P, Hapgood KP, Morton DAV. Bulk Flow Optimisation of Amorphous Solid Dispersion Excipient Powders through Surface Modification. Pharmaceutics 2023; 15:pharmaceutics15051447. [PMID: 37242689 DOI: 10.3390/pharmaceutics15051447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Particulate amorphous solid dispersions (ASDs) have been recognised for their potential to enhance the performance of various solid dose forms, especially oral bioavailability and macromolecule stability. However, the inherent nature of spray-dried ASDs leads to their surface cohesion/adhesion, including hygroscopicity, which hinders their bulk flow and affects their utility and viability in terms of powder production, processing, and function. This study explores the effectiveness of L-leucine (L-leu) coprocessing in modifying the particle surface of ASD-forming materials. Various contrasting prototype coprocessed ASD excipients from both the food and pharmaceutical industries were examined for their effective coformulation with L-leu. The model/prototype materials included maltodextrin, polyvinylpyrrolidone (PVP K10 and K90), trehalose, gum arabic, and hydroxypropyl methylcellulose (HPMC E5LV and K100M). The spray-drying conditions were set such that the particle size difference was minimised, so that it did not play a substantial role in influencing powder cohesion. Scanning electron microscopy was used to evaluate the morphology of each formulation. A combination of previously reported morphological progression typical of L-leu surface modification and previously unreported physical characteristics was observed. The bulk characteristics of these powders were assessed using a powder rheometer to evaluate their flowability under confined and unconfined stresses, flow rate sensitivities, and compactability. The data showed a general improvement in maltodextrin, PVP K10, trehalose and gum arabic flowability measures as L-leu concentrations increased. In contrast, PVP K90 and HPMC formulations experienced unique challenges that provided insight into the mechanistic behaviour of L-leu. Therefore, this study recommends further investigations into the interplay between L-leu and the physico-chemical properties of coformulated excipients in future amorphous powder design. This also revealed the need to enhance bulk characterisation tools to unpack the multifactorial impact of L-leu surface modification.
Collapse
Affiliation(s)
- Danni Suhaidi
- School of Engineering, Deakin University, Waurn Ponds, VIC 3216, Australia
| | - Yao-Da Dong
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Paul Wynne
- Medicines Manufacturing Innovation Centre, Monash University, Clayton, VIC 3168, Australia
| | - Karen P Hapgood
- School of Engineering, Swinburne University, Hawthorn, VIC 3122, Australia
| | - David A V Morton
- School of Engineering, Deakin University, Waurn Ponds, VIC 3216, Australia
| |
Collapse
|
21
|
Kulkarni VR, Chakka J, Alkadi F, Maniruzzaman M. Veering to a Continuous Platform of Fused Deposition Modeling Based 3D Printing for Pharmaceutical Dosage Forms: Understanding the Effect of Layer Orientation on Formulation Performance. Pharmaceutics 2023; 15:pharmaceutics15051324. [PMID: 37242565 DOI: 10.3390/pharmaceutics15051324] [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: 03/10/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Three-dimensional (3D) printing of pharmaceuticals has been centered around the idea of personalized patient-based 'on-demand' medication. Fused deposition modeling (FDM)-based 3D printing processes provide the capability to create complex geometrical dosage forms. However, the current FDM-based processes are associated with printing lag time and manual interventions. The current study tried to resolve this issue by utilizing the dynamic z-axis to continuously print drug-loaded printlets. Fenofibrate (FNB) was formulated with hydroxypropyl methylcellulose (HPMC AS LG) into an amorphous solid dispersion using the hot-melt extrusion (HME) process. Thermal and solid-state analyses were used to confirm the amorphous state of the drug in both polymeric filaments and printlets. Printlets with a 25, 50, and 75% infill density were printed using the two printing systems, i.e., continuous, and conventional batch FDM printing methods. Differences between the two methods were observed in the breaking force required to break the printlets, and these differences reduced as the infill density went up. The effect on in vitro release was significant at lower infill densities but reduced at higher infill densities. The results obtained from this study can be used to understand the formulation and process control strategies when switching from conventional FDM to the continuous printing of 3D-printed dosage forms.
Collapse
Affiliation(s)
- Vineet R Kulkarni
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78705, USA
| | - Jaidev Chakka
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78705, USA
| | - Faez Alkadi
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78705, USA
| | - Mohammed Maniruzzaman
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78705, USA
| |
Collapse
|
22
|
Du Y, Struppe J, Perrone B, Hassan A, Codina A, Su Y. Efficient analysis of pharmaceutical drug substances and products using a solid-state NMR CryoProbe. Analyst 2023; 148:724-734. [PMID: 36722866 DOI: 10.1039/d2an01903e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Solid-state nuclear magnetic resonance (ssNMR) is a high-resolution and versatile spectroscopic tool for characterizing pharmaceutical solids. However, the inherent low sensitivity of NMR remains a significant challenge in the analysis of natural abundance drug substances and products. Here, we report, for the first time, the application of a CPMAS CryoProbe™ to improve the sensitivity of 13C and 15N detection by approximately 5 to 6 times for solid-state analysis of a commercial pharmaceutical drug posaconazole (POSA). The sensitivity enhancement enables two-dimensional (2D) 13C-13C and 1H-15N correlation experiments, which are otherwise time-prohibitive using regular MAS probes, for resonance assignment and structural elucidation. These polarization transfer and correlation experiments reveal drug-drug and drug-polymer interactions in amorphous POSA and its amorphous solid dispersion formulation. Our results demonstrated that the CPMAS CryoProbe™ can be widely applied for routine pharmaceutical analysis and advanced structural investigations with significantly enhanced efficiency and throughput.
Collapse
Affiliation(s)
- Yong Du
- Analytical Research & Development, Merck & Co., Inc., Rahway, NJ 07065, USA.
| | | | | | - Alia Hassan
- Bruker Switzerland AG, 8117 Faellanden, Switzerland
| | | | - Yongchao Su
- Analytical Research & Development, Merck & Co., Inc., Rahway, NJ 07065, USA.
| |
Collapse
|
23
|
Stegemann S, Moreton C, Svanbäck S, Box K, Motte G, Paudel A. Trends in oral small-molecule drug discovery and product development based on product launches before and after the Rule of Five. Drug Discov Today 2023; 28:103344. [PMID: 36442594 DOI: 10.1016/j.drudis.2022.103344] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/28/2022] [Accepted: 09/01/2022] [Indexed: 11/26/2022]
Abstract
In 1997, the 'Rule of Five' (Ro5) suggested physicochemical limitations for orally administered drugs, based on the analysis of chemical libraries from the early 1990s. In this review, we report on the trends in oral drug product development by analyzing products launched between 1994 and 1997 and between 2013 and 2019. Our analysis confirmed that most new oral drugs are within the Ro5 descriptors; however, the number of new drug products of drugs with molecular weight (MW) and calculated partition coefficient (clogP) beyond the Ro5 has slightly increased. Analysis revealed that there is no single scientific or technological reason for this trend, but that it likely results from incremental advances are being made in molecular biology, target diversity, drug design, medicinal chemistry, predictive modeling, drug metabolism and pharmacokinetics, and drug delivery.
Collapse
Affiliation(s)
- Sven Stegemann
- Institute for Process and Particle Engineering, Graz University of Technology, Inffeldgasse 13, 8010 Graz, Austria.
| | | | - Sami Svanbäck
- The Solubility Company Ltd, Viikinkaari 4, 00790 Helsinki, Finland
| | - Karl Box
- Pion Inc. (UK) Ltd, Forest Row, UK
| | - Geneviève Motte
- JEN Pharma Consulting, 182 Rue Henri Latour, 1450 Chastre, Belgium
| | - Amrit Paudel
- Institute for Process and Particle Engineering, Graz University of Technology, Inffeldgasse 13, 8010 Graz, Austria; Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria
| |
Collapse
|
24
|
Pugliese A, Tobyn M, Hawarden LE, Abraham A, Blanc F. New Development in Understanding Drug-Polymer Interactions in Pharmaceutical Amorphous Solid Dispersions from Solid-State Nuclear Magnetic Resonance. Mol Pharm 2022; 19:3685-3699. [PMID: 36037249 PMCID: PMC9644399 DOI: 10.1021/acs.molpharmaceut.2c00479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 01/08/2023]
Abstract
Pharmaceutical amorphous solid dispersions (ASDs) represent a widely used technology to increase the bioavailability of active pharmaceutical ingredients (APIs). ASDs are based on an amorphous API dispersed in a polymer, and their stability is driven by the presence of strong intermolecular interactions between these two species (e.g., hydrogen bond, electrostatic interactions, etc.). The understanding of these interactions at the atomic level is therefore crucial, and solid-state nuclear magnetic resonance (NMR) has demonstrated itself as a very powerful technique for probing API-polymer interactions. Other reviews have also reported exciting approaches to study the structures and dynamic properties of ASDs and largely focused on the study of API-polymer miscibility and on the identification of API-polymer interactions. Considering the increased use of NMR in the field, the aim of this Review is to specifically highlight recent experimental strategies used to identify API-polymer interactions and report promising recent examples using one-dimensional (1D) and two-dimensional (2D) experiments by exploiting the following emerging approaches of very-high magnetic field and ultrafast magic angle spinning (MAS). A range of different ASDs spanning APIs and polymers with varied structural motifs is targeted to illustrate new ways to understand the mechanism of stability of ASDs to enable the design of new dispersions.
Collapse
Affiliation(s)
- Andrea Pugliese
- Department
of Chemistry, University of Liverpool, Liverpool L69 7ZD, United Kingdom
| | - Michael Tobyn
- Drug
Product Development, Bristol-Myers Squibb, Moreton CH46 1QW, United Kingdom
| | - Lucy E. Hawarden
- Drug
Product Development, Bristol-Myers Squibb, Moreton CH46 1QW, United Kingdom
| | - Anuji Abraham
- Drug
Product Development, Bristol-Myers Squibb, New Brunswick, New Jersey 08903, United States
| | - Frédéric Blanc
- Department
of Chemistry, University of Liverpool, Liverpool L69 7ZD, United Kingdom
- Stephenson
Institute for Renewable Energy, University
of Liverpool, Liverpool L69 7ZF, United Kingdom
| |
Collapse
|
25
|
Deon M, dos Santos J, de Andrade DF, Beck RCR. A critical review of traditional and advanced characterisation tools to drive formulators towards the rational development of 3D printed oral dosage forms. Int J Pharm 2022; 628:122293. [DOI: 10.1016/j.ijpharm.2022.122293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 10/03/2022] [Accepted: 10/09/2022] [Indexed: 10/31/2022]
|
26
|
Wang X, Zhu Y, Zhao X, Zhang S, Cao M, Wang X, Li W. Development and characterization of an amorphous Curcumin-Eudragit®E100 Solid Dispersions with improved solubility, stability, and pharmacokinetic properties. Pharm Dev Technol 2022; 27:965-974. [DOI: 10.1080/10837450.2022.2141778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Affiliation(s)
- Xin Wang
- Department of Medicinal Chemistry, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Yijian Zhu
- Department of Medicinal Chemistry, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Xudong Zhao
- Department of Medicinal Chemistry, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Shurong Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Meiting Cao
- Department of Medicinal Chemistry, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoyue Wang
- Department of Medicinal Chemistry, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
27
|
Recent Advances in Amorphous Solid Dispersions: Preformulation, Formulation Strategies, Technological Advancements and Characterization. Pharmaceutics 2022; 14:pharmaceutics14102203. [PMID: 36297638 PMCID: PMC9609913 DOI: 10.3390/pharmaceutics14102203] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/27/2022] [Accepted: 10/13/2022] [Indexed: 11/05/2022] Open
Abstract
Amorphous solid dispersions (ASDs) are among the most popular and widely studied solubility enhancement techniques. Since their inception in the early 1960s, the formulation development of ASDs has undergone tremendous progress. For instance, the method of preparing ASDs evolved from solvent-based approaches to solvent-free methods such as hot melt extrusion and Kinetisol®. The formulation approaches have advanced from employing a single polymeric carrier to multiple carriers with plasticizers to improve the stability and performance of ASDs. Major excipient manufacturers recognized the potential of ASDs and began introducing specialty excipients ideal for formulating ASDs. In addition to traditional techniques such as differential scanning calorimeter (DSC) and X-ray crystallography, recent innovations such as nano-tomography, transmission electron microscopy (TEM), atomic force microscopy (AFM), and X-ray microscopy support a better understanding of the microstructure of ASDs. The purpose of this review is to highlight the recent advancements in the field of ASDs with respect to formulation approaches, methods of preparation, and advanced characterization techniques.
Collapse
|
28
|
Paladino E, Doerr FJ, Bordos E, Onyemelukwe II, Lamprou DA, Florence AJ, Gilmore IS, Halbert GW. High spatial resolution ToF-SIMS imaging and image analysis strategies to monitor and quantify early phase separation in amorphous solid dispersions. Int J Pharm 2022; 628:122191. [DOI: 10.1016/j.ijpharm.2022.122191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/18/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022]
|
29
|
Melt Fusion Techniques for Solubility Enhancement: A Comparison of Hot Melt Extrusion and KinetiSol® Technologies. Sci Pharm 2022. [DOI: 10.3390/scipharm90030051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
A successful candidate for oral drug delivery needs to possess adequate solubility and dissolution rate to elicit its therapeutic action. Extensive research is being carried out to enhance the solubility of poorly soluble drugs through a number of techniques involving polymeric and non-polymeric approaches. Non-polymeric approaches such as micronization and nanocrystals are successful in improving the apparent solubility of drugs, but the sustenance of solubility is not always possible. Amorphous solid dispersions (ASDs) lead to solubility enhancement as well as the maintenance of solubility with the assistance of polymers, thereby improving bioavailability. Spray drying, hot melt extrusion (HME), and KinetiSol® technologies are some of the techniques capable of manufacturing ASDs. Each of these techniques has its own advantages and disadvantages in terms of processing challenges and applicability in preparing ASDs. The latter two technologies are similar in being fusion and non-solvent techniques to improve solubility. This review compares both HME and KinetiSol® techniques regarding mechanism, equipment design, formulation, and process parameters involved and scalability.
Collapse
|
30
|
Effects of Polymers on the Drug Solubility and Dissolution Enhancement of Poorly Water-Soluble Rivaroxaban. Int J Mol Sci 2022; 23:ijms23169491. [PMID: 36012748 PMCID: PMC9409000 DOI: 10.3390/ijms23169491] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/15/2022] [Accepted: 08/20/2022] [Indexed: 01/12/2023] Open
Abstract
The purpose of this study was to investigate the efficacy of hydrophilic polymers in a solid dispersion formulation in improving the solubility and dissolution rate of rivaroxaban (RXB), a poorly soluble drug. The developed solid dispersion consisted of two components, a drug and a polymer, and the drug was dispersed as amorphous particles in a polymer matrix using the spray drying method. Polymeric solid dispersions were evaluated using solubility tests, in vitro dissolution tests, powder X-ray diffraction, differential scanning calorimetry, scanning electron microscopy, and particle size distribution analysis. To maximize physical stability against crystallization and improve the solubility and dissolution of RXB, it is important to select the appropriate polymer type and the optimal ratio of the polymer to the drug. The optimized polyvinyl alcohol (PVA)-based (1/0.5, w/w) and gelatin-based (1/5, w/w) solid dispersion formulations showed 6.3 and 3.6 times higher drug solubilities than pure RXB powder, respectively, and the final dissolution rate was improved by approximately 1.5 times. Scanning electron microscopy and particle size distribution analyses confirmed that the gelatin-based solid dispersion was smaller and more spherical than the PVA-based solid dispersion, suggesting that the gelatin-based solid dispersion had a faster initial dissolution rate. Differential scanning calorimetry and powder X-ray diffraction analyses confirmed that RXB had successfully changed from a crystalline form to an amorphous form, contributing to the improvement in its solubility and dissolution rate. This study provides a strategy for selecting suitable polymers for the development of amorphous polymer solid dispersions that can overcome precipitation during dissolution and stabilization of the amorphous state. In addition, the selected polymer solid dispersion improved the drug solubility and dissolution rate of RXB, a poorly soluble drug, and may be used as a promising drug delivery system.
Collapse
|
31
|
Du Y, Su Y. 19F Solid-state NMR characterization of pharmaceutical solids. SOLID STATE NUCLEAR MAGNETIC RESONANCE 2022; 120:101796. [PMID: 35688018 DOI: 10.1016/j.ssnmr.2022.101796] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
Solid-state NMR has been increasingly recognized as a high-resolution and versatile spectroscopic tool to characterize drug substances and products. However, the analysis of pharmaceutical materials is often carried out at natural isotopic abundance and a relatively low drug loading in multi-component systems and therefore suffers from challenges of low sensitivity. The fact that fluorinated therapeutics are well represented in pipeline drugs and commercial products offers an excellent opportunity to utilize fluorine as a molecular probe for pharmaceutical analysis. We aim to review recent advancements of 19F magic angle spinning NMR methods in modern drug research and development. Applications to polymorph screening at the micromolar level, structural elucidation, and investigation of molecular interactions at the Ångström to submicron resolution in drug delivery, stability, and quality will be discussed.
Collapse
Affiliation(s)
- Yong Du
- Analytical Research and Development, Merck & Co., Inc., Rahway, NJ, 07065, United States
| | - Yongchao Su
- Analytical Research and Development, Merck & Co., Inc., Rahway, NJ, 07065, United States; Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, IN, 47907, United States; Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, United States; Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT, 06269, United States.
| |
Collapse
|
32
|
Downstream Processing of Itraconazole:HPMCAS Amorphous Solid Dispersion: From Hot-Melt Extrudate to Tablet Using a Quality by Design Approach. Pharmaceutics 2022; 14:pharmaceutics14071429. [PMID: 35890324 PMCID: PMC9323274 DOI: 10.3390/pharmaceutics14071429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/17/2022] Open
Abstract
The downstream processing of hot-melt extruded amorphous solid dispersions (ASDs) into tablets is challenging due to the low tabletability of milled ASDs. Typically, the extrudate strand is sized before milling, as the strand cannot be fed directly into the milling system. At the lab scale, the strand can be sized by hand-cutting before milling. For scaling up, pelletizers or chill roll and flaker systems can be used to break strands. Due to the different techniques used, differences in milling and tablet compaction are to be expected. We present a systematic study of the milling and tableting of an extruded ASD of itraconazole with hypromellose acetate succinate (HPMCAS) as a carrier polymer. The strand was sized using different techniques at the end of the extruder barrel (hand-cutting, pelletizer, or chill roll and flaker) before being milled at varying milling speeds with varying screen sizes. The effects of these variables (sizing technology, milling speed, and screen size) on the critical quality attributes (CQAs) of the milled ASD, such as yield, mean particle size (D50), tablet compaction characteristics, and tablet dissolution, were established using response surface methodology. It was found that the CQAs varied according to sizing technology, with chill roll flakes showing the highest percentage yield, the lowest D50, and the highest tabletability and dissolution rate for itraconazole. Pearson correlation coefficient tests indicated D50 as the most important CQA related to tabletability and dissolution. For certain milling conditions, the milling of hand-cut filaments results in similar particle size distributions (PSDs) to the milling of pellets or chill roll flakes.
Collapse
|
33
|
Školáková T, Smržová D, Pekárek T, Lhotka M, Školáková A, Klimša V, Kadeřábková A, Zámostný P. Investigation of tadalafil molecular arrangement in solid dispersions using inverse gas chromatography and Raman mapping. Int J Pharm 2022; 623:121955. [PMID: 35753537 DOI: 10.1016/j.ijpharm.2022.121955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/06/2022] [Accepted: 06/21/2022] [Indexed: 11/30/2022]
Abstract
The aim of this study was to investigate the molecular structures of tadalafil solid dispersions prepared by different techniques and further to relate them to surface free energy information indicating the final amorphousness of the product. Thus, we tried to complement the existing knowledge of solid dispersion formation. Poorly water-soluble tadalafil was combined with different polymers, i.e. Kollidon® 12 PF, Kollidon® VA 64 and Soluplus®, to form model systems. To assess the extent of drug-polymer miscibility, we studied model solid dispersion surface energy using inverse gas chromatography and phase micro-structure using confocal Raman microscopy. The selection of the preparation method was found to play a crucial role in the molecular arrangement of the incorporated drug and the polymer in resulting solid dispersion. Our results showed that a lower surface free energy indicated the formation of a more homogeneous solid dispersion. Conversely, a higher surface free energy corresponded to the heterogeneous systems containing tadalafil amorphous clusters that were captured by Raman mapping. Thus, we successfully introduced a novel evaluation approach of the drug molecular arrangement in solid dispersions that is especially useful for examining the miscibility of the components when the conventional characterizing techniques are inconclusive or yield variable results.
Collapse
Affiliation(s)
- Tereza Školáková
- Department of Organic Technology, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic.
| | - Dominika Smržová
- Department of Organic Technology, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Tomáš Pekárek
- Zentiva, k.s., U Kabelovny 130, 102 37 Prague 10, Czech Republic
| | - Miloslav Lhotka
- Department of Inorganic Technology, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Andrea Školáková
- Department of Metals and Corrosion Engineering, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Vojtěch Klimša
- Department of Chemical Engineering, University of Chemistry and Technology Prague, Technická 3, 166 28 Prague 6, Czech Republic
| | - Alena Kadeřábková
- Department of Polymers, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Petr Zámostný
- Department of Organic Technology, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| |
Collapse
|
34
|
Santitewagun S, Thakkar R, Zeitler JA, Maniruzzaman M. Detecting Crystallinity Using Terahertz Spectroscopy in 3D Printed Amorphous Solid Dispersions. Mol Pharm 2022; 19:2380-2389. [PMID: 35670498 DOI: 10.1021/acs.molpharmaceut.2c00163] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
This study demonstrates the applicability of terahertz time-domain spectroscopy (THz-TDS) in evaluating the solid-state of the drug in selective laser sintering-based 3D printed dosage forms. Selective laser sintering is a powder bed-based 3D printing platform, which has recently demonstrated applicability in manufacturing amorphous solid dispersions (ASDs) through a layer-by-layer fusion process. When formulating ASDs, it is critical to confirm the final solid state of the drug as residual crystallinity can alter the performance of the formulation. Moreover, SLS 3D printing does not involve the mixing of the components during the process, which can lead to partially amorphous systems causing reproducibility and storage stability problems along with possibilities of unwanted polymorphism. In this study, a previously investigated SLS 3D printed ASD was characterized using THz-TDS and compared with traditionally used solid-state characterization techniques, including differential scanning calorimetry (DSC) and powder X-ray diffractometry (pXRD). THz-TDS provided deeper insights into the solid state of the dosage forms and their properties. Moreover, THz-TDS was able to detect residual crystallinity in granules prepared using twin-screw granulation for the 3D printing process, which was undetectable by the DSC and XRD. THz-TDS can prove to be a useful tool in gaining deeper insights into the solid-state properties and further aid in predicting the stability of amorphous solid dispersions.
Collapse
Affiliation(s)
- Supawan Santitewagun
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, U.K
| | - Rishi Thakkar
- Pharmaceutical Engineering and 3D printing Lab (PharmE3D), The Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| | - J Axel Zeitler
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, U.K
| | - Mohammed Maniruzzaman
- Pharmaceutical Engineering and 3D printing Lab (PharmE3D), The Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
35
|
Amorphous characterization of pharmaceutical drug substances enabled by the elastic modulus mapping of atomic force microscope. Int J Pharm 2022; 621:121784. [DOI: 10.1016/j.ijpharm.2022.121784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 11/19/2022]
|
36
|
Solid Dispersion Formulations by FDM 3D Printing-A Review. Pharmaceutics 2022; 14:pharmaceutics14040690. [PMID: 35456524 PMCID: PMC9032529 DOI: 10.3390/pharmaceutics14040690] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 01/06/2023] Open
Abstract
Additive manufacturing (AM) is revolutionizing the way medicines are designed, manufactured, and utilized. Perhaps, AM appears to be ideal for the fit-for-purpose manufacturing of medicines in contrast to the several disadvantages associated with the conventional fit-for-all mass production that accounts for less than 50% of pharmacotherapeutic treatment/management of diseases especially among children and elderly patients, as well as patients with special needs. In this review, we discuss the current trends in the application of additive manufacturing to prepare personalized dosage forms on-demand focusing the attention on the relevance of coupling solid dispersion with FDM 3D printing. Combining the two technologies could offer many advantages such as to improve the solubility, dissolution, and oral bioavailability of poorly soluble drugs in tandem with the concept of precision medicine and personalized dosing and to address the dilemma of commercial availability of FDM filaments loaded with Class II and/or Class IV drugs. However, thermal treatment especially for heat-sensitive drugs, regulatory, and ethical obligations in terms of quality control and quality assurance remain points of concern. Hence, a concerted effort is needed between the scientific community, the pharmaceutical industries, the regulatory agencies, the clinicians and clinical pharmacists, and the end-users to address these concerns.
Collapse
|
37
|
Stegemann S, Faulhammer E, Pinto JT, Paudel A. Focusing on powder processing in dry powder inhalation product development, manufacturing and performance. Int J Pharm 2022; 614:121445. [PMID: 34998921 DOI: 10.1016/j.ijpharm.2021.121445] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/17/2021] [Accepted: 12/31/2021] [Indexed: 12/12/2022]
Abstract
Dry powder inhalers (DPI) are well established products for the delivery of actives via the pulmonary route. Various DPI products are marketed or developed for the treatment of local lung diseases such as chronic obstructive pulmonary disease (COPD), asthma or cystic fibrosis as well as systemic diseases targeted through inhaled delivery (i.e. Diabetes Mellitus). One of the key prerequisites of DPI formulations is that the aerodynamic size of the drug particles needs to be below 5 µm to enter deeply into the respiratory tract. These inherently cohesive inhalable size particles are either formulated as adhesive mixture with coarse carrier particles like lactose called carrier-based DPI or are formulated as free-flowing carrier-free particles (e.g. soft agglomerates, large hollow particles). In either case, it is common practice that drug and/or excipient particles of DPI formulations are obtained by processing API and API/excipients. The DPI manufacturing process heavily involves several particle and powder technologies such as micronization of the API, dry blending, powder filling and other particle engineering processes such as spray drying, crystallization etc. In this context, it is essential to thoroughly understand the impact of powder/particle properties and processing on the quality and performance of the DPI formulations. This will enable prediction of the processability of the DPI formulations and controlling the manufacturing process so that meticulously designed formulations are able to be finally developed as the finished DPI dosage form. This article is intended to provide a concise account of various aspects of DPI powder processing, including the process understanding and material properties that are important to achieve the desired DPI product quality. Various endeavors of model informed formulation/process design and development for DPI powder and PAT enabled process monitoring and control are also discussed.
Collapse
Affiliation(s)
- Sven Stegemann
- Institute of Process and Particle Engineering, Graz University of Technology, Inffeldgasse 13, 8010 Graz, Austria
| | - Eva Faulhammer
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria
| | - Joana T Pinto
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria
| | - Amrit Paudel
- Institute of Process and Particle Engineering, Graz University of Technology, Inffeldgasse 13, 8010 Graz, Austria; Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria.
| |
Collapse
|
38
|
Khan KU, Minhas MU, Badshah SF, Suhail M, Ahmad A, Ijaz S. Overview of nanoparticulate strategies for solubility enhancement of poorly soluble drugs. Life Sci 2022; 291:120301. [PMID: 34999114 DOI: 10.1016/j.lfs.2022.120301] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/27/2021] [Accepted: 01/01/2022] [Indexed: 12/20/2022]
Abstract
Poor aqueous solubility and poor bioavailability are major issues with many pharmaceutical industries. By some estimation, 70-90% drug candidates in development stage while up-to 40% of the marketed products are poorly soluble which leads to low bioavailability, reduced therapeutic effects and dosage escalation. That's why solubility is an important factor to consider during design and manufacturing of the pharmaceutical products. To-date, various strategies have been explored to tackle the issue of poor solubility. This review article focuses the updated overview of commonly used macro and nano drug delivery systems and techniques such as micronization, solid dispersion (SD), supercritical fluid (SCF), hydrotropy, co-solvency, micellar solubilization, cryogenic technique, inclusion complex formation-based techniques, nanosuspension, solid lipid nanoparticles, and nanogels/nanomatrices explored for solubility enhancement of poorly soluble drugs. Among various techniques, nanomatrices were found a promising and impeccable strategy for solubility enhancement of poorly soluble drugs. This article also describes the mechanism of action of each technique used in solubilization enhancement.
Collapse
Affiliation(s)
- Kifayat Ullah Khan
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, 63100, Punjab, Pakistan; Quaid-e-Azam College of Pharmacy, Sahiwal, Punjab, Pakistan
| | - Muhammad Usman Minhas
- College of Pharmacy, University of Sargodha, University Road, Sargodha City, Punjab, Pakistan.
| | - Syed Faisal Badshah
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, 63100, Punjab, Pakistan
| | - Muhammad Suhail
- School of Pharmacy, Kaohsiung Medical University, 100 Shih-Chuan Ist Road, Kaohsiung City 807, Taiwan, ROC
| | - Aousaf Ahmad
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, 63100, Punjab, Pakistan; Quaid-e-Azam College of Pharmacy, Sahiwal, Punjab, Pakistan
| | - Shakeel Ijaz
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, 63100, Punjab, Pakistan; Quaid-e-Azam College of Pharmacy, Sahiwal, Punjab, Pakistan
| |
Collapse
|
39
|
Thompson SA, Davis DA, Moon C, Williams RO. Increasing Drug Loading of Weakly Acidic Telmisartan in Amorphous Solid Dispersions through pH Modification during Hot-Melt Extrusion. Mol Pharm 2022; 19:318-331. [PMID: 34846902 DOI: 10.1021/acs.molpharmaceut.1c00805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Oral drug therapy requiring large quantities of active pharmaceutical ingredients (APIs) can cause a substantial pill burden, which can increase nonadherence and worsen healthcare outcomes. Maximizing the drug loading of APIs in oral dosage forms is essential to reduce pill burden. This can be challenging for poorly water-soluble APIs without compromising performance. We show a promising strategy for maximizing the drug loading of pH-dependent APIs in amorphous solid dispersions (ASDs) produced by hot-melt extrusion (HME) without compromising their dissolution performance. We examine potential increases in the drug loading (w/w) of telmisartan in ASDs by incorporating bases to modify pH during HME. Telmisartan is a weakly acidic, poorly water-soluble API with pH-dependent solubility. It is practically insoluble at physiological pH, but its solubility increases exponentially at pH values above 10. Telmisartan was extruded with the polymer Soluplus and various bases. With no base, the maximum drug loading achieved by extrusion was only 5% before crystalline telmisartan was detected. Including a strong, water-soluble base (NaOH or KOH) increased the maximum amorphous drug loading to 50%. These results indicate that telmisartan has pH-dependent solubility in a molten polymer, similar to that in an aqueous solution. We also examine the stability of Soluplus when extruded with a strong base, using solid-state nuclear magnetic resonance (ssNMR) to determine that NaOH (but not KOH) causes degradation by hydrolysis. Supersaturation was maintained for at least 20 h during dissolution testing of a 50% telmisartan ASD in biorelevant media.
Collapse
Affiliation(s)
- Stephen A Thompson
- Division of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin College of Pharmacy, Austin, Texas 78712, United States
| | - Daniel A Davis
- Division of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin College of Pharmacy, Austin, Texas 78712, United States
| | - Chaeho Moon
- Division of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin College of Pharmacy, Austin, Texas 78712, United States
| | - Robert O Williams
- Division of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin College of Pharmacy, Austin, Texas 78712, United States
| |
Collapse
|
40
|
Chen Z, Higashi K, Ueda K, Moribe K. Transition from Amorphous Cyclosporin A Nanoparticles to Size-Reduced Stable Nanocrystals in a Poloxamer 407 Solution. Mol Pharm 2022; 19:188-199. [PMID: 34843257 DOI: 10.1021/acs.molpharmaceut.1c00721] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Amorphous drug nanoparticles usually exhibit low storage stability. A comprehensive understanding of the molecular states and physicochemical properties of the product is indispensable for designing stable formulations. In the present study, an amorphous cyclosporin A (CyA) nanosuspension with a mean particle size of approximately 370 nm was prepared by wet bead milling with poloxamer 407 (P407). Interestingly, the prepared amorphous CyA nanoparticles were transformed into uniform CyA nanocrystals with a reduced mean particle size of approximately 200 nm during storage at 25 °C. The CyA nanocrystals were stably maintained for at least 1 month. The particle morphologies and molecular structures of the CyA nanosuspensions before and after storage were thoroughly characterized by cryogenic transmission electron microscopy and magic-angle spinning nuclear magnetic resonance spectroscopy, respectively. They revealed that the freshly prepared amorphous CyA nanoparticles (∼370 nm) were secondary particles composed of aggregated primary particles with an estimated size of 50 nm. A portion of P407 was found to be entrapped at the gaps between the primary particles due to aggregation, while most of P407 was dissolved in the solution either adsorbing at the solid/liquid interface or forming polymeric micelles. The entrapped P407 is considered to play an important role in the destabilization of the amorphous CyA nanoparticles. The resultant CyA nanocrystals (∼200 nm) were uniform single crystals of Form 2 hydrate and showed corner-truncated bipyramidal features. Owing to the narrow particle size distribution of the CyA nanocrystals, the rate of Ostwald ripening was slow, giving long-term stability to the CyA nanocrystals. This study provides new insights into the destabilization mechanism of amorphous drug nanoparticles.
Collapse
Affiliation(s)
- Ziqiao Chen
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Kenjirou Higashi
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Keisuke Ueda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Kunikazu Moribe
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| |
Collapse
|
41
|
Zhang Q, Durig T, Blass B, Fassihi R. Development of an amorphous based sustained release system for apremilast a selective phosphodiesterase 4 (PDE4) inhibitor. Int J Pharm 2022; 615:121516. [DOI: 10.1016/j.ijpharm.2022.121516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 10/19/2022]
|
42
|
Characterization of Novel Solid Dispersions of Moringa oleifera Leaf Powder Using Thermo-Analytical Techniques. Processes (Basel) 2021. [DOI: 10.3390/pr9122230] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Moringa oleifera leaf powder (MOLP) has been identified as the most important functional ingredient owing to its rich nutritional profile and healthy effects. The solubility and functional properties of this ingredient can be enhanced through solid dispersion technology. This study aimed to investigate the effects of polyethylene glycols (PEGs) 4000 and 6000 as hydrophilic carriers and solid dispersion techniques (freeze-drying, melting, solvent evaporation, and microwave irradiation) on the crystallinity and thermal stability of solid-dispersed Moringa oleifera leaf powders (SDMOLPs). SDMOLPs were dully characterized using powder X-ray diffraction (PXRD), differential scanning calorimetry (DSC), thermo-gravimetric analysis (TGA), and Fourier transform infrared spectroscopy (FTIR). The PXRD results revealed that the solid dispersions were partially amorphous with strong diffraction peaks at 2θ values of 19° and 23°. The calorimetric and thermogravimetric curves showed that PEGs conferred greater stability on the dispersions. The FTIR studyrevealed the existence of strong intermolecular hydrogen bond interactions between MOLP and PEG functional groups. MOLP solid dispersions may be useful in functional foods and beverages and nutraceutical formulations.
Collapse
|
43
|
Kapourani A, Tzakri T, Valkanioti V, Kontogiannopoulos KN, Barmpalexis P. Drug crystal growth in ternary amorphous solid dispersions: Effect of surfactants and polymeric matrix-carriers. Int J Pharm X 2021; 3:100086. [PMID: 34151251 PMCID: PMC8193146 DOI: 10.1016/j.ijpx.2021.100086] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 01/10/2023] Open
Abstract
The present study evaluates the crystal growth rate of amorphous drugs when dispersed in different ternary polymeric amorphous solid dispersions (ASDs) in the presence of surfactants. Specifically, ternary ASDs of aprepitant (APT, selected as a model drug) were prepared via melt-quench cooling by evaluating three commonly used ASDs matrix/carriers, namely hydroxypropyl cellulose (HPC), poly(vinylpyrrolidone) (PVP) and the copolymer Soluplus® (SOL), and two suitable surfactants, namely d-alpha tocopheryl polyethylene glycol 1000 succinate (TPGS) and poly(ethylene glycol)-block-poly(propylene glycol)-block-poly(ethylene glycol) (P407). Results showed that all components were completely miscible (verified via hot stage polarized microscopy) and both surfactants were acting as plasticizers to the API. APT's crystal growth rate was increased in the presence of both P407 and TPGS, while PVP was identified as the matrix/carrier with the greatest impact API's crystal growth rate inhibition. Interestingly, TPGS presented a noticeable synergistic effect when combined with PVP resulting in a further reduction of APT's crystal growth rate. Furthermore, evaluation of APT's nucleation induction time in dissolution medium (PBS pH 6.8) revealed PVP as the most effective crystallization inhibitor, whereas the addition of TPGS showed to improve PVP's ability to inhibit APT's recrystallization. Finally, the formation of intermolecular interactions in the ternary APT-PVP-TPGS provided an explanation for the observed PVP-TPGS synergistic effects, with molecular dynamics simulations being able to unravel the type and extent of these interactions on a theoretical basis.
Collapse
Affiliation(s)
- Afroditi Kapourani
- Department of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Theodora Tzakri
- Department of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Vasiliki Valkanioti
- Department of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Konstantinos N. Kontogiannopoulos
- Department of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
- Natural Products Research Centre of Excellence-AUTH (NatPro-AUTH), Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Thessaloniki 57001, Greece
| | - Panagiotis Barmpalexis
- Department of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
- Natural Products Research Centre of Excellence-AUTH (NatPro-AUTH), Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Thessaloniki 57001, Greece
| |
Collapse
|
44
|
Quodbach J, Bogdahn M, Breitkreutz J, Chamberlain R, Eggenreich K, Elia AG, Gottschalk N, Gunkel-Grabole G, Hoffmann L, Kapote D, Kipping T, Klinken S, Loose F, Marquetant T, Windolf H, Geißler S, Spitz T. Quality of FDM 3D Printed Medicines for Pediatrics: Considerations for Formulation Development, Filament Extrusion, Printing Process and Printer Design. Ther Innov Regul Sci 2021; 56:910-928. [PMID: 34826120 PMCID: PMC9492703 DOI: 10.1007/s43441-021-00354-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/04/2021] [Indexed: 02/08/2023]
Abstract
3d printing is capable of providing dose individualization for pediatric medicines and translating the precision medicine approach into practical application. In pediatrics, dose individualization and preparation of small dosage forms is a requirement for successful therapy, which is frequently not possible due to the lack of suitable dosage forms. For precision medicine, individual characteristics of patients are considered for the selection of the best possible API in the most suitable dose with the most effective release profile to improve therapeutic outcome. 3d printing is inherently suitable for manufacturing of individualized medicines with varying dosages, sizes, release profiles and drug combinations in small batch sizes, which cannot be manufactured with traditional technologies. However, understanding of critical quality attributes and process parameters still needs to be significantly improved for this new technology. To ensure health and safety of patients, cleaning and process validation needs to be established. Additionally, adequate analytical methods for the in-process control of intermediates, regarding their printability as well as control of the final 3d printed tablets considering any risk of this new technology will be required. The PolyPrint consortium is actively working on developing novel polymers for fused deposition modeling (FDM) 3d printing, filament formulation and manufacturing development as well as optimization of the printing process, and the design of a GMP-capable FDM 3d printer. In this manuscript, the consortium shares its views on quality aspects and measures for 3d printing from drug-loaded filaments, including formulation development, the printing process, and the printed dosage forms. Additionally, engineering approaches for quality assurance during the printing process and for the final dosage form will be presented together with considerations for a GMP-capable printer design.
Collapse
Affiliation(s)
- Julian Quodbach
- Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany.
| | - Malte Bogdahn
- Merck Healthcare KGaA, Frankfurter Str. 250, Darmstadt, Germany
| | - Jörg Breitkreutz
- Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Rebecca Chamberlain
- Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | | | | | | | | | - Lena Hoffmann
- Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | | | - Thomas Kipping
- Merck Life Science KGaA, Frankfurter Str. 250, Darmstadt, Germany
| | - Stefan Klinken
- Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Fabian Loose
- Laboratory for Manufacturing Systems, University of Applied Sciences Cologne, Betzdorfer Str. 2, 50679, Cologne, Germany
| | | | - Hellen Windolf
- Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Simon Geißler
- Merck Healthcare KGaA, Frankfurter Str. 250, Darmstadt, Germany
| | - Tilmann Spitz
- Laboratory for Manufacturing Systems, University of Applied Sciences Cologne, Betzdorfer Str. 2, 50679, Cologne, Germany
| |
Collapse
|
45
|
Mansuri A, Münzner P, Feuerbach T, Vermeer AWP, Hoheisel W, Böhmer R, Thommes M, Gainaru C. The relaxation behavior of supercooled and glassy imidacloprid. J Chem Phys 2021; 155:174502. [PMID: 34742219 DOI: 10.1063/5.0067404] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Employing dielectric spectroscopy, oscillatory shear rheology, and calorimetry, the present work explores the molecular dynamics of the widely used insecticide imidacloprid above and below its glass transition temperature. In its supercooled liquid regime, the applied techniques yield good agreement regarding the characteristic structural (alpha) relaxation times of this material. In addition, the generalized Gemant-DiMarzio-Bishop model provides a good conversion between the frequency-dependent dielectric and shear mechanical responses in its viscous state, allowing for an assessment of imidacloprid's molecular hydrodynamic radius. In order to characterize the molecular dynamics in its glassy regime, we employ several approaches. These include the application of frequency-temperature superposition (FTS) to its isostructural dielectric and rheological responses as well as use of dielectric and calorimetric physical aging and the Adam-Gibbs-Vogel model. While the latter approach and dielectric FTS provide relaxation times that are close to each other, the other methods predict notably longer times that are closer to those reflecting a complete recovery of ergodicity. This seemingly conflicting dissimilarity demonstrates that the molecular dynamics of glassy imidacloprid strongly depends on its thermal history, with high relevance for the use of this insecticide as an active ingredient in technological applications.
Collapse
Affiliation(s)
- A Mansuri
- INVITE GmbH, 51368 Leverkusen, Germany
| | - P Münzner
- Department of Physics, TU Dortmund University, 44221 Dortmund, Germany
| | - T Feuerbach
- Chair of Solids Process Engineering, TU Dortmund University, 44227 Dortmund, Germany
| | | | | | - R Böhmer
- Department of Physics, TU Dortmund University, 44221 Dortmund, Germany
| | - M Thommes
- Chair of Solids Process Engineering, TU Dortmund University, 44227 Dortmund, Germany
| | - C Gainaru
- Department of Physics, TU Dortmund University, 44221 Dortmund, Germany
| |
Collapse
|
46
|
Iyer R, Petrovska Jovanovska V, Berginc K, Jaklič M, Fabiani F, Harlacher C, Huzjak T, Sanchez-Felix MV. Amorphous Solid Dispersions (ASDs): The Influence of Material Properties, Manufacturing Processes and Analytical Technologies in Drug Product Development. Pharmaceutics 2021; 13:1682. [PMID: 34683975 PMCID: PMC8540358 DOI: 10.3390/pharmaceutics13101682] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/06/2021] [Accepted: 09/13/2021] [Indexed: 12/14/2022] Open
Abstract
Poorly water-soluble drugs pose a significant challenge to developability due to poor oral absorption leading to poor bioavailability. Several approaches exist that improve the oral absorption of such compounds by enhancing the aqueous solubility and/or dissolution rate of the drug. These include chemical modifications such as salts, co-crystals or prodrugs and physical modifications such as complexation, nanocrystals or conversion to amorphous form. Among these formulation strategies, the conversion to amorphous form has been successfully deployed across the pharmaceutical industry, accounting for approximately 30% of the marketed products that require solubility enhancement and making it the most frequently used technology from 2000 to 2020. This article discusses the underlying scientific theory and influence of the active compound, the material properties and manufacturing processes on the selection and design of amorphous solid dispersion (ASD) products as marketed products. Recent advances in the analytical tools to characterize ASDs stability and ability to be processed into suitable, patient-centric dosage forms are also described. The unmet need and regulatory path for the development of novel ASD polymers is finally discussed, including a description of the experimental data that can be used to establish if a new polymer offers sufficient differentiation from the established polymers to warrant advancement.
Collapse
Affiliation(s)
- Raman Iyer
- Technical Research and Development, c/o Global Drug Development, Novartis Pharmaceuticals Corp., One Health Plaza, East Hanover, NJ 07936, USA
| | - Vesna Petrovska Jovanovska
- Product Development, Lek Pharmaceuticals d.d., Verovškova 57, 1526 Ljubljana, Slovenia; (V.P.J.); (K.B.); (M.J.); (T.H.)
| | - Katja Berginc
- Product Development, Lek Pharmaceuticals d.d., Verovškova 57, 1526 Ljubljana, Slovenia; (V.P.J.); (K.B.); (M.J.); (T.H.)
| | - Miha Jaklič
- Product Development, Lek Pharmaceuticals d.d., Verovškova 57, 1526 Ljubljana, Slovenia; (V.P.J.); (K.B.); (M.J.); (T.H.)
| | - Flavio Fabiani
- Technical Research and Development, c/o Global Drug Development, Novartis Pharma AG, Lichtstrasse 35, CH-4056 Basel, Switzerland; (F.F.); (C.H.)
| | - Cornelius Harlacher
- Technical Research and Development, c/o Global Drug Development, Novartis Pharma AG, Lichtstrasse 35, CH-4056 Basel, Switzerland; (F.F.); (C.H.)
| | - Tilen Huzjak
- Product Development, Lek Pharmaceuticals d.d., Verovškova 57, 1526 Ljubljana, Slovenia; (V.P.J.); (K.B.); (M.J.); (T.H.)
| | | |
Collapse
|
47
|
Jiang X, Zhao Y, Guan Q, Xiao S, Dong W, Lian S, Zhang H, Liu M, Wang Z, Han J. Amorphous solid dispersions of cyclosporine A with improved bioavailability prepared via hot melt extrusion: Formulation, physicochemical characterization, and in vivo evaluation. Eur J Pharm Sci 2021; 168:106036. [PMID: 34637896 DOI: 10.1016/j.ejps.2021.106036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/08/2021] [Accepted: 09/02/2021] [Indexed: 02/05/2023]
Abstract
In this study, the amorphous solid dispersions of cyclosporine A (CsA-ASDs) were prepared by hot melt extrusion (HME) with PVP K12 as carrier to improve the oral bioavailability of CsA. The polymers were screened by solubilization and recrystallization inhibition experiments, then the CsA-ASDs were prepared with optimized technological parameters and characterized on thermodynamics and morphology. The results showed that CsA was dispersed among PVP K12 as amorphous form in CsA-ASDs, and the infrared spectrum testified that there was possible hydrogen bond interaction between CsA and PVP K12. The in vivo pharmacokinetics of CsA formulations in rats were analyzed via LC-MS. The AUC of CsA-ASD tablets increased by 7.3 times compared to CsA bulk powder and 3.1 times in contrast to CsA-PM tablets, respectively. The experiment proved that CsA-ASD tablets significantly improved the dissolution and absorption of the drug. This study had a reference value for the bioavailability improvement of oral CsA preparations.
Collapse
Affiliation(s)
- Xinxin Jiang
- Institute of Biopharmaceutical Research, Liaocheng University, Hunan Road, Liaocheng, Shandong 252059, People's Republic of China
| | - Yanna Zhao
- Institute of Biopharmaceutical Research, Liaocheng University, Hunan Road, Liaocheng, Shandong 252059, People's Republic of China.
| | - Qingran Guan
- Institute of Biopharmaceutical Research, Liaocheng University, Hunan Road, Liaocheng, Shandong 252059, People's Republic of China
| | - Shanshan Xiao
- Institute of Biopharmaceutical Research, Liaocheng University, Hunan Road, Liaocheng, Shandong 252059, People's Republic of China
| | - Weimiao Dong
- Institute of Biopharmaceutical Research, Liaocheng University, Hunan Road, Liaocheng, Shandong 252059, People's Republic of China
| | - Shipeng Lian
- Shandong Weifang Rainbow Chemical Co., Ltd, Weifang, Shandong 261100, People's Republic of China
| | - Huaizhen Zhang
- School of Environment and Planning, Liaocheng University, Hunan Road, Liaocheng, Shandong 252059, People's Republic of China
| | - Min Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Hunan Road, Liaocheng, Shandong 252059, People's Republic of China; School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng, Shandong 252059, People's Republic of China
| | - Zhengping Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Hunan Road, Liaocheng, Shandong 252059, People's Republic of China; Liaocheng High-Tech Biotechnology Co. Ltd, Liaocheng, Shandong 252059, People's Republic of China
| | - Jun Han
- Institute of Biopharmaceutical Research, Liaocheng University, Hunan Road, Liaocheng, Shandong 252059, People's Republic of China; Liaocheng High-Tech Biotechnology Co. Ltd, Liaocheng, Shandong 252059, People's Republic of China.
| |
Collapse
|
48
|
Tsiaxerli A, Karagianni A, Ouranidis A, Kachrimanis K. Polyelectrolyte Matrices in the Modulation of Intermolecular Electrostatic Interactions for Amorphous Solid Dispersions: A Comprehensive Review. Pharmaceutics 2021; 13:pharmaceutics13091467. [PMID: 34575543 PMCID: PMC8468962 DOI: 10.3390/pharmaceutics13091467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/04/2021] [Accepted: 09/09/2021] [Indexed: 12/05/2022] Open
Abstract
Polyelectrolyte polymers have been widely used in the pharmaceutical field as excipients to facilitate various drug delivery systems. Polyelectrolytes have been used to modulate the electrostatic environment and enhance favorable interactions between the drug and the polymer in amorphous solid dispersions (ASDs) prepared mainly by hot-melt extrusion. Polyelectrolytes have been used alone, or in combination with nonionic polymers as interpolyelectrolyte complexes, or after the addition of small molecular additives. They were found to enhance physical stability by favoring stabilizing intermolecular interactions, as well as to exert an antiplasticizing effect. Moreover, they not only enhance drug dissolution, but they have also been used for maintaining supersaturation, especially in the case of weakly basic drugs that tend to precipitate in the intestine. Additional uses include controlled and/or targeted drug release with enhanced physical stability and ease of preparation via novel continuous processes. Polyelectrolyte matrices, used along with scalable manufacturing methods in accordance with green chemistry principles, emerge as an attractive viable alternative for the preparation of ASDs with improved physical stability and biopharmaceutic performance.
Collapse
Affiliation(s)
- Anastasia Tsiaxerli
- Department of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.T.); (A.K.); (A.O.)
| | - Anna Karagianni
- Department of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.T.); (A.K.); (A.O.)
| | - Andreas Ouranidis
- Department of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.T.); (A.K.); (A.O.)
- Department of Chemical Engineering, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Kyriakos Kachrimanis
- Department of Pharmaceutical Technology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.T.); (A.K.); (A.O.)
- Correspondence: ; Tel.: +30-2310-997666
| |
Collapse
|
49
|
Hempel NJ, Knopp MM, Zeitler JA, Berthelsen R, Löbmann K. Microwave-Induced in Situ Drug Amorphization Using a Mixture of Polyethylene Glycol and Polyvinylpyrrolidone. J Pharm Sci 2021; 110:3221-3229. [PMID: 34022194 DOI: 10.1016/j.xphs.2021.05.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/13/2021] [Accepted: 05/13/2021] [Indexed: 01/03/2023]
Abstract
The use of a mixture of polyethylene glycol (PEG) and polyvinylpyrrolidone (PVP) was investigated for microwave-induced in situ amorphization of celecoxib (CCX) inside compacts. Such amorphization requires the presence of a dipolar excipient in the formulation to ensure heating of the compact by absorption of the microwaves. Previously, the hygroscopic nature of PVP was exploited for this purpose. By exposing PVP-based compacts for set time intervals at defined relative humidity, controlled water sorption into the compacts was achieved. In the present study, PEG was proposed as the microwave absorbing excipient instead of water, to avoid the water sorption step. However, it was found that PEG alone melted upon exposure to microwave radiation and caused the compact to deform. Furthermore, CCX was found to recrystallize upon cooling in PEG-based formulations. Hence, a mixture of PEG and PVP was used, where the presence of PVP preserved the physical shape of the compact, and the physical state of the amorphous solid dispersion. To study the impact of the polymer mixture, different compact compositions of CCX, PEG and PVP were prepared. When exposing the compacts to microwave radiation, it was found that the PEG:PVP ratio was critical for in situ amorphization and that complete amorphization was only achieved above a certain temperature threshold.
Collapse
Affiliation(s)
| | | | - J Axel Zeitler
- Department of Chemical Engineering, University of Cambridge, Cambridge CB3 0AS, UK
| | - Ragna Berthelsen
- Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Korbinian Löbmann
- Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark.
| |
Collapse
|
50
|
Borde S, Paul SK, Chauhan H. Ternary solid dispersions: classification and formulation considerations. Drug Dev Ind Pharm 2021; 47:1011-1028. [PMID: 33818224 DOI: 10.1080/03639045.2021.1908342] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The number of active pharmaceutical compounds from the biopharmaceutical classification system (BCS) belonging to Class II and IV have significantly increased in recent years. These compounds have high therapeutic potential but are difficult to formulate as oral dosage forms due to their poor aqueous solubility. The solubility and bioavailability of these poorly water-soluble compounds can be increased by various formulation approaches, such as amorphous solid dispersions (ASD), salt formation, complexations, etc. Out of these techniques, the ASD approach, where compounds are converted into amorphous form and embedded in the hydrophilic matrix, have been successfully used in many marketed preparations. The recent advancement of this ASD approach is the design of ternary solid dispersions (TSD), where an additional component is added to further improve their performance in terms of solubility, stability, and processability. This review discusses the classification, mechanism of performance improvement, preparation techniques, and characterizations for TSD.
Collapse
Affiliation(s)
- Shambhavi Borde
- School of Pharmacy and Health Professions, Creighton University, Omaha, NE, USA
| | - Sagar Kumar Paul
- School of Pharmacy and Health Professions, Creighton University, Omaha, NE, USA
| | - Harsh Chauhan
- School of Pharmacy and Health Professions, Creighton University, Omaha, NE, USA
| |
Collapse
|