1
|
Walter S, Mileo PGM, Afzal MAF, Kyeremateng SO, Degenhardt M, Browning AR, Shelley JC. Predicting the Release Mechanism of Amorphous Solid Dispersions: A Combination of Thermodynamic Modeling and In Silico Molecular Simulation. Pharmaceutics 2024; 16:1292. [PMID: 39458621 PMCID: PMC11510624 DOI: 10.3390/pharmaceutics16101292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/18/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND During the dissolution of amorphous solid dispersion (ASD) formulations, the drug load (DL) often impacts the release mechanism and the occurrence of loss of release (LoR). The ASD/water interfacial gel layer and its specific phase behavior in connection with DL strongly dictate the release mechanism and LoR of ASDs, as reported in the literature. Thermodynamically driven liquid-liquid phase separation (LLPS) and/or drug crystallization at the interface are the key phase transformations that drive LoR. METHODS In this study, a combination of Perturbed-Chain Statistical Associating Fluid Theory (PC-SAFT) thermodynamic modeling and in silico molecular simulation was applied to investigate the release mechanism and the occurrence LoR of an ASD formulation consisting of ritonavir as the active pharmaceutical ingredient (API) and the polymer, polyvinylpyrrolidone-co-vinyl acetate (PVPVA64). A thermodynamically modeled ternary phase diagram of ritonavir (PVPVA64) and water was applied to predict DL-dependent LLPS in the ASD/water interfacial gel layer. Microscopic Erosion Time Testing (METT) was used to experimentally validate the phase diagram predictions. Additionally, in silico molecular simulation was applied to provide further insights into the phase separation, the release mechanism, and aggregation behavior on a molecular level. RESULTS Thermodynamic modeling, molecular simulation, and experimental results were consistent and complementary, providing evidence that ASD/water interactions and phase separation are essential factors driving the dissolution behavior and LoR at 40 wt% DL of the investigated ritonavir/PVPVA64 ASD system, consistent with previous studies. CONCLUSIONS This study provides insights into the potential of blending thermodynamic modeling, molecular simulation, and experimental research to comprehensively understand ASD formulations. Such a combined approach can be leveraged as a computational framework to gain insights into the ASD dissolution mechanism, thereby facilitating in silico screening, designing, and optimization of formulations with the benefit of significantly reducing the number of experimental tests.
Collapse
Affiliation(s)
- Stefanie Walter
- AbbVie Deutschland GmbH & Co. KG, Small Molecule CMC Development, Knollstraße, 67061 Ludwigshafen am Rhein, Germany; (S.W.); (M.D.)
| | - Paulo G. M. Mileo
- Materials Science, Schrödinger GmbH, Glücksteinallee 25, 68163 Mannheim, Germany;
| | - Mohammad Atif Faiz Afzal
- Materials Science, Schrödinger LLC, Suite 1300, 101 SW Main Street, Portland, OR 97204, USA; (M.A.F.A.); (A.R.B.)
| | - Samuel O. Kyeremateng
- AbbVie Deutschland GmbH & Co. KG, Small Molecule CMC Development, Knollstraße, 67061 Ludwigshafen am Rhein, Germany; (S.W.); (M.D.)
| | - Matthias Degenhardt
- AbbVie Deutschland GmbH & Co. KG, Small Molecule CMC Development, Knollstraße, 67061 Ludwigshafen am Rhein, Germany; (S.W.); (M.D.)
| | - Andrea R. Browning
- Materials Science, Schrödinger LLC, Suite 1300, 101 SW Main Street, Portland, OR 97204, USA; (M.A.F.A.); (A.R.B.)
| | - John C. Shelley
- Life Science, Schrödinger LLC, Suite 1300, 101 SW Main Street, Portland, OR 97204, USA
| |
Collapse
|
2
|
Larsen BS, Meiland P, Tzdaka E, Tho I, Rades T. A unifying approach to drug-in-polymer solubility prediction: Streamlining experimental workflow and analysis. Eur J Pharm Biopharm 2024; 203:114478. [PMID: 39226986 DOI: 10.1016/j.ejpb.2024.114478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/05/2024]
Abstract
This method paper describes currently used experimental methods to predict the drug-in-polymer solubility of amorphous solid dispersions and offers a combined approach for applying the Melting-point-depression method, the Recrystallization method, and the Melting-and-mixing method. It aims to describe and expand on the theoretical basis as well as the analytical methodology of the recently published Melting-and-mixing method. This solubility method relies on determining the relationship between drug loads and the enthalpy of melting and mixing of a crystalline drug in the presence of an amorphous polymer. This relationship is used to determine the soluble drug load of an amorphous solid dispersion from the recorded enthalpy of melting and mixing of the crystalline drug portion in a drug-polymer sample at equilibrium solubility. Due to the complex analytical methodology of the Melting-and-mixing method, a software solution called the Glass Solution Companion app was developed. Using this new tool, it is possible to calculate the predicted drug-in-polymer solubility and Flory-Huggins interaction parameter from experimental samples, as well as to generate the resulting solubility-temperature curve. This software can be used for calculations for all three experimental methods, which would be useful for comparing the applicability of the methods on a given drug-polymer system. Since it is difficult to predict the suitability of these drug-in-polymer solubility methods for a specific drug-polymer system in silico, some experimental investigation is necessary. By optimizing the experimental protocol, it is possible to collect data for the three experimental methods simultaneously for a specific drug-polymer system. These results can then be readily analyzed using the Glass Solution Companion app to find the most appropriate method for the drug-polymer system, and therefore, the most reliable drug-in-polymer solubility prediction.
Collapse
Affiliation(s)
- Bjarke Strøm Larsen
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Sem Sælands vei 3, 0371 Oslo, Norway.
| | - Peter Meiland
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark
| | - Eidan Tzdaka
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Sem Sælands vei 3, 0371 Oslo, Norway
| | - Ingunn Tho
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Sem Sælands vei 3, 0371 Oslo, Norway.
| | - Thomas Rades
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| |
Collapse
|
3
|
Antolović I, Vrabec J, Klajmon M. COSMOPharm: Drug-Polymer Compatibility of Pharmaceutical Amorphous Solid Dispersions from COSMO-SAC. Mol Pharm 2024; 21:4395-4415. [PMID: 39078049 PMCID: PMC11372840 DOI: 10.1021/acs.molpharmaceut.4c00342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
The quantum mechanics-aided COSMO-SAC activity coefficient model is applied and systematically examined for predicting the thermodynamic compatibility of drugs and polymers. The drug-polymer compatibility is a key aspect in the rational selection of optimal polymeric carriers for pharmaceutical amorphous solid dispersions (ASD) that enhance drug bioavailability. The drug-polymer compatibility is evaluated in terms of both solubility and miscibility, calculated using standard thermodynamic equilibrium relations based on the activity coefficients predicted by COSMO-SAC. As inherent to COSMO-SAC, our approach relies only on quantum-mechanically derived σ-profiles of the considered molecular species and involves no parameter fitting to experimental data. All σ-profiles used were determined in this work, with those of the polymers being derived from their shorter oligomers by replicating the properties of their central monomer unit(s). Quantitatively, COSMO-SAC achieved an overall average absolute deviation of 13% in weight fraction drug solubility predictions compared to experimental data. Qualitatively, COSMO-SAC correctly categorized different polymer types in terms of their compatibility with drugs and provided meaningful estimations of the amorphous-amorphous phase separation. Furthermore, we analyzed the sensitivity of the COSMO-SAC results for ASD to different model configurations and σ-profiles of polymers. In general, while the free volume and dispersion terms exerted a limited effect on predictions, the structures of oligomers used to produce σ-profiles of polymers appeared to be more important, especially in the case of strongly interacting polymers. Explanations for these observations are provided. COSMO-SAC proved to be an efficient method for compatibility prediction and polymer screening in ASD, particularly in terms of its performance-cost ratio, as it relies only on first-principles calculations for the considered molecular species. The open-source nature of both COSMO-SAC and the Python-based tool COSMOPharm, developed in this work for predicting the API-polymer thermodynamic compatibility, invites interested readers to explore and utilize this method for further research or assistance in the design of pharmaceutical formulations.
Collapse
Affiliation(s)
- Ivan Antolović
- Thermodynamics, Technische Universität Berlin, Ernst-Reuter-Platz 1, 10587 Berlin, Germany
| | - Jadran Vrabec
- Thermodynamics, Technische Universität Berlin, Ernst-Reuter-Platz 1, 10587 Berlin, Germany
| | - Martin Klajmon
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czechia
| |
Collapse
|
4
|
Wünsche S, Tenberg V, Oliynyk K, Seidel-Morgenstern A, Lorenz H, Sadeghi M. Experimental and model-based approach to evaluate solvent effects on the solubility of the pharmaceutical artemisinin. Eur J Pharm Sci 2024; 200:106826. [PMID: 38866112 DOI: 10.1016/j.ejps.2024.106826] [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: 04/16/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/14/2024]
Abstract
The separation and purification of plant-based Active Pharmaceutical Ingredients (API) from extracts is a crucial part in pharmaceutical process development. For the purification of the antimalarial drug component artemisinin (ARTE) from an Artemisia anna L. toluene extract, antisolvent crystallization is considered. Solubilities of ARTE in binary solvent mixtures of toluene and two potential antisolvents, n-heptane and ethanol, were determined at temperatures from 278.15 K to 313.15 K. The experimental work was supported by the application of various models, utilizing varying amounts of experimental input data. The goal was the identification of models that are able to predict solubilities in binary solvent mixtures sufficiently accurate and, thus, can help to reduce the experimental effort for future solvent screenings. In this study, we applied the PC-SAFT model both with and without fitting the binary interaction parameter kij between ARTE and the respective solvent, as well as the empirical Jouyban-Acree model. From the experiments, n-heptane demonstrated to be a promising antisolvent, while ethanol acted more as a cosolvent. All models tested were capable of distinguishing between effective and ineffective antisolvents. The purely predictive PC-SAFT model applied with kij = 0 exhibited the largest deviation from the experimental data. This was followed by the PC-SAFT model including fitted kij values, based on at least four experimental data points. The Jouyban-Acree model fitted the data most accurately. Its parametrization required a minimum of ten experimental data points.
Collapse
Affiliation(s)
- Steffi Wünsche
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Vico Tenberg
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Karina Oliynyk
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | | | - Heike Lorenz
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
| | - Masoud Sadeghi
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany.
| |
Collapse
|
5
|
Deac A, Luebbert C, Qi Q, Courtney RM, Indulkar AS, Gao Y, Zhang GGZ, Sadowski G, Taylor LS. Dissolution Mechanisms of Amorphous Solid Dispersions: Application of Ternary Phase Diagrams To Explain Release Behavior. Mol Pharm 2024; 21:1900-1918. [PMID: 38469754 DOI: 10.1021/acs.molpharmaceut.3c01179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
The use of amorphous solid dispersions (ASDs) in commercial drug products has increased in recent years due to the large number of poorly soluble drugs in the pharmaceutical pipeline. However, the release behavior of ASDs is complex and remains not well understood. Often, the drug release from ASDs is rapid and complete at lower drug loadings (DLs) but becomes slow and incomplete at higher DLs. The DL where release becomes hindered is termed the limit of congruency (LoC). Currently, there are no approaches to predict the LoC. However, recent findings show that one potential cause leading to the LoC is a change in phase morphology after water-induced phase separation at the ASD/solution interface. In this study, the phase behavior of ASDs in contact with aqueous solutions was described thermodynamically by constructing experimental and computational ternary phase diagrams, and these were used to predict morphology changes and ultimately the LoC. Experimental ternary phase diagrams were obtained by equilibrating ASD/water mixtures over time. Computational ternary phase diagrams were obtained by Perturbed Chain Statistical Associating Fluid Theory (PC-SAFT). The morphology of the hydrophobic phase was studied with fluorescence confocal microscopy. It was demonstrated that critical point (plait point) composition approximately corresponded to the ASD DL, where the hydrophobic phase, formed during phase separation, became interconnected and hindered ASD release. This work provides mechanistic insights into the ASD release behavior and highlights the potential of in silico ASD design using phase diagrams.
Collapse
Affiliation(s)
- Alexandru Deac
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | | | - Qingqing Qi
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Reagan M Courtney
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Anura S Indulkar
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Yi Gao
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Geoff G Z Zhang
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | | | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
6
|
Casalini T, Mann J, Pepin X. Predicting Surface pH in Unbuffered Conditions for Acids, Bases, and Their Salts - A Review of Modeling Approaches and Their Performance. Mol Pharm 2024; 21:513-534. [PMID: 38127789 DOI: 10.1021/acs.molpharmaceut.3c00661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Dissolution of ionizable drugs and their salts is a function of drug surface solubility driven by the surface pH, i.e., the microenvironmental pH at the solid/liquid interface, which will deviate from bulk pH when there is an acid-base reaction occurring at the solid/liquid interface. In this work, we first present a brief overview of the modeling approaches available in the literature, classified according to the rate-determining step assumed in the dissolution process. In the second part, we present and evaluate the prediction performance of two different modeling approaches for surface pH. The first method relies only on thermodynamic equilibria, while the second method accounts for transport phenomena of charged compounds through the diffusional boundary layer using the Nernst - Planck equation. Model outcomes are compared with experimental data taken from the literature and obtained during this work. In terms of surface pH predictions, the models provide identical values for weak acids or weak bases. The models' outcomes for bases are in good agreement with experimental data in acidic conditions (bulk pH 1-4), while overpredictions are observed in the 5-7 bulk pH range in a system-dependent manner. Deviations can be related to the effect of surface dissolution (also referred to as surface reaction), which may become a controlling mechanism and slow the replenishment of the unionized drug at the surface of the crystal. Surface pH predictions for acids are generally in good agreement with experiments, with a slight underestimation for some drug examples, which could be related to errors in intrinsic solubility determination or to the assumption of thermodynamic equilibrium at the surface of the drug. A good agreement is also observed for salts with the thermodynamic model except for mesylate salts, suggesting that other phenomena, not currently included in the thermodynamic equilibrium model, may determine the surface pH.
Collapse
Affiliation(s)
- Tommaso Casalini
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg 431 50, Sweden
| | - James Mann
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield SK10 2NA, U.K
| | - Xavier Pepin
- New Modalities & Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield SK10 2NA, U.K
| |
Collapse
|
7
|
Iemtsev A, Zumaya ALV, Dinh M, Hassouna F, Fulem M. Towards rational design of API-poly(D, L-lactide-co-glycolide) based micro- and nanoparticles: The role of API-polymer compatibility prediction. Int J Pharm 2024; 650:123724. [PMID: 38123107 DOI: 10.1016/j.ijpharm.2023.123724] [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: 08/14/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 12/23/2023]
Abstract
Due to their unique properties, such as controlled drug release and improved bioavailability, polymeric microparticles and nanoparticles (MPs and NPs) have gained considerable interest in the pharmaceutical industry. Nevertheless, the high costs associated with biodegradable polymers and the active pharmaceutical ingredients (APIs) used for treating serious diseases, coupled with the vast number of API-polymer combinations, make the search for effective API-polymer MPs and NPs a costly and time-consuming process. In this work, the correlation between the compatibility of selected model APIs (i.e., ibuprofen, naproxen, paracetamol, and indomethacin) with poly(lactide-co-glycolide) (PLGA) derived from respective binary phase diagrams and characteristics of prepared MPs and NPs, such as the drug loading and solid-state properties, was investigated to probe the possibility of implementing the modeling of API-polymer thermodynamic and kinetic phase behavior as part of rational design of drug delivery systems based on MPs and NPs. API-PLGA-based MPs and NPs were formulated using an emulsion-solvent evaporation technique and were characterized for morphology, mean size, zeta potential, drug loading, and encapsulation efficiency. The solid-state properties of the encapsulated APIs were assessed using differential scanning calorimetry and X-ray powder diffraction. The evaluated compatibility was poor for all considered API-PLGA pairs, which is in alignment with the experimental results showing low drug loading in terms of amorphous API content. At the same time, drug loading of the studied APIs in terms of amorphous content was found to follow the same trend as their solubility in PLGA, indicating a clear correlation between API solubility in PLGA and achievable drug loading. These findings suggest that API-polymer phase behavior modeling and compatibility screening can be employed as an effective preformulation tool to estimate optimum initial API concentration for MP and NP preparation or, from a broader perspective, to tune or select polymeric carriers offering desired drug loading.
Collapse
Affiliation(s)
- Anton Iemtsev
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Alma Lucia Villela Zumaya
- Faculty of Chemical Engineering, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Martin Dinh
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Fatima Hassouna
- Faculty of Chemical Engineering, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic.
| | - Michal Fulem
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic.
| |
Collapse
|
8
|
Pisay M, Padya S, Mutalik S, Koteshwara KB. Stability Challenges of Amorphous Solid Dispersions of Drugs: A Critical Review on Mechanistic Aspects. Crit Rev Ther Drug Carrier Syst 2024; 41:45-94. [PMID: 38037820 DOI: 10.1615/critrevtherdrugcarriersyst.2023039877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
The most common drawback of the existing and novel drug molecules is their low bioavailability because of their low solubility. One of the most important approaches to enhance the bioavailability in the enteral route for poorly hydrophilic molecules is amorphous solid dispersion (ASD). The solubility of compounds in amorphous form is comparatively high because of the availability of free energy produced during formulation. This free energy results in the change of crystalline nature of the prepared ASD to the stable crystalline form leading to the reduced solubility of the product. Due to the intrinsic chemical and physical uncertainty and the restricted knowledge about the interactions of active molecules with the carriers making, this ASD is a challenging task. This review focused on strategies to stabilize ASD by considering the various theories explaining the free-energy concept, physical interactions, and thermal properties. This review also highlighted molecular modeling and machine learning computational advancement to stabilize ASD.
Collapse
Affiliation(s)
- Muralidhar Pisay
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Singh Padya
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Kunnatur B Koteshwara
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| |
Collapse
|
9
|
Mathers A, Pechar M, Hassouna F, Fulem M. The step-wise dissolution method: An efficient DSC-based protocol for verification of predicted API-polymer compatibility. Int J Pharm 2023; 648:123604. [PMID: 37981251 DOI: 10.1016/j.ijpharm.2023.123604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/03/2023] [Accepted: 11/11/2023] [Indexed: 11/21/2023]
Abstract
The development of an amorphous solid dispersion (ASD) is a promising strategy for improving the low bioavailability of many poorly water-soluble active pharmaceutical ingredients (APIs). The construction of a temperature-composition (T-C) phase diagram for an API-polymer combination is imperative as it can provide critical information that is essential for formulating stable ASDs. However, the currently followed differential scanning calorimetry (DSC)-based strategies for API solubility determination in a polymer at elevated temperatures are inefficient and, on occasions, unreliable, which may lead to an inaccurate prediction at lower temperatures of interest (i.e., T = 25 °C). Recently, we proposed a novel DSC-based protocol called the "step-wise dissolution" (S-WD) method, which is both cost- and time-effective. The objective of this study was to test the applicability of the S-WD method regarding expeditious verification of the purely-predicted API-polymer compatibility via the perturbed chain-statistical associating fluid theory (PC-SAFT) equation of state (EOS). Fifteen API-polymer T-C phase diagrams were reliably constructed, with three distinct API-polymer case types being identified regarding the approach used for the S-WD method. Overall, the PC-SAFT EOS provided satisfactory qualitative descriptions of the API-polymer compatibility, but not necessarily accurate quantitative predictions of the API solubility in the polymer at T = 25 °C. The S-WD method was subsequently modified and an optimal protocol was proposed, which can significantly reduce the required experimental effort.
Collapse
Affiliation(s)
- Alex Mathers
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Matouš Pechar
- Faculty of Chemical Engineering, University of Chemistry and Technology, Prague, Technická 3, 166 28 Prague 6, Czech Republic
| | - Fatima Hassouna
- Faculty of Chemical Engineering, University of Chemistry and Technology, Prague, Technická 3, 166 28 Prague 6, Czech Republic
| | - Michal Fulem
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic.
| |
Collapse
|
10
|
Luebbert C, Stoyanov E. Tailored ASD destabilization - Balancing shelf life stability and dissolution performance with hydroxypropyl cellulose. Int J Pharm X 2023; 5:100187. [PMID: 37396620 PMCID: PMC10314205 DOI: 10.1016/j.ijpx.2023.100187] [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: 02/28/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 07/04/2023] Open
Abstract
Amorphous solid dispersion (ASD) formulations are preferred enabling formulations for poorly water soluble active pharmaceutical ingredients (API) as they reliably enhance the dissolution behavior and solubility. Balancing a high stability against unwanted transformations such as crystallization and amorphous phase separation during storage on the one hand and optimizing the dissolution behavior of the formulation (high supersaturation and maintenance for long time) on the other hand are essential during formulation development. This study assessed the potential of ternary ASDs (one API and two polymers) containing the polymers hydroxypropyl cellulose together with poly(vinylpyrrolidone-co-vinyl acetate) (PVP VA64) or hydroxypropyl cellulose acetate succinate to stabilize the amorphously embedded APIs fenofibrate and simvastatin during storage and to enhance the dissolution performance. Thermodynamic predictions using the PC-SAFT model revealed for each combination of polymers the optimal polymer ratio, maximum API load that is thermodynamically stable as well as miscibility of the two polymers. The stability predictions were validated by three months enduring stability tests, followed by a characterization of the dissolution behavior. The thermodynamically most stable ASDs were found to be the ASDs with deteriorated dissolution performance. Within the investigated polymer combinations, physical stability and dissolution performance opposed each other.
Collapse
Affiliation(s)
| | - Edmont Stoyanov
- Nisso Chemical Europe GmbH, Berliner Allee 42, Düsseldorf D-40212, Germany
| |
Collapse
|
11
|
Klueppelberg J, Handge UA, Thommes M, Winck J. Composition Dependency of the Flory-Huggins Interaction Parameter in Drug-Polymer Phase Behavior. Pharmaceutics 2023; 15:2650. [PMID: 38139992 PMCID: PMC10747291 DOI: 10.3390/pharmaceutics15122650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 12/24/2023] Open
Abstract
An innovative strategy to address recent challenges in the oral administration of poorly soluble drugs is the formulation of amorphous solid dispersions (ASDs), where the drug is dissolved in a highly soluble carrier polymer. Therefore, special knowledge of the drug-polymer phase behavior is essential for an effective product and process design, accelerating the introduction of novel efficacious ASD products. Flory-Huggins theory can be applied to model solubility temperatures of crystalline drugs in carrier polymers over the drug fraction. However, predicted solubility temperatures lack accuracy in cases of strong drug/polymer interactions that are not represented in the Flory-Huggins lattice model. Within this study, a modeling strategy is proposed to improve the predictive power through an extension of the Flory-Huggins interaction parameter by a correlation with the drug fraction. Therefore, the composition dependency of the Flory-Huggins interaction parameter was evaluated experimentally for various drug-polymer formulations that cover a wide variety of drug and polymer characteristics regarding molecular weights, glass transition temperatures and melting temperatures, as well as drug-polymer interactions of different strengths and effects. The extended model was successfully approved for nine exemplary ASD formulations containing the drugs acetaminophen, itraconazole, and griseofulvine, as well as the following polymers: basic butylated methacrylate copolymer, Soluplus®, and vinylpyrrolidone/vinyl acetate copolymer. A high correlation between the predicted solubility temperatures and experimental and literature data was found, particularly at low drug fractions, since the model accounts for composition dependent drug-polymer interactions.
Collapse
Affiliation(s)
- Jana Klueppelberg
- Laboratory of Solids Process Engineering, Department of Biochemical and Chemical Engineering, TU Dortmund University, Emil-Figge-Street 68, 44227 Dortmund, Germany; (J.K.); (M.T.)
| | - Ulrich A. Handge
- Chair of Plastics Technology, Department of Mechanical Engineering, TU Dortmund University, Leonhard-Euler-Street 5, 44227 Dortmund, Germany;
| | - Markus Thommes
- Laboratory of Solids Process Engineering, Department of Biochemical and Chemical Engineering, TU Dortmund University, Emil-Figge-Street 68, 44227 Dortmund, Germany; (J.K.); (M.T.)
| | - Judith Winck
- Laboratory of Solids Process Engineering, Department of Biochemical and Chemical Engineering, TU Dortmund University, Emil-Figge-Street 68, 44227 Dortmund, Germany; (J.K.); (M.T.)
| |
Collapse
|
12
|
Pavliš J, Mathers A, Fulem M, Klajmon M. Can Pure Predictions of Activity Coefficients from PC-SAFT Assist Drug-Polymer Compatibility Screening? Mol Pharm 2023; 20:3960-3974. [PMID: 37386723 PMCID: PMC10410664 DOI: 10.1021/acs.molpharmaceut.3c00124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 07/01/2023]
Abstract
The bioavailability of poorly water-soluble active pharmaceutical ingredients (APIs) can be improved via the formulation of an amorphous solid dispersion (ASD), where the API is incorporated into a suitable polymeric carrier. Optimal carriers that exhibit good compatibility (i.e., solubility and miscibility) with given APIs are typically identified through experimental means, which are routinely labor- and cost-inefficient. Therefore, the perturbed-chain statistical associating fluid theory (PC-SAFT) equation of state, a popular thermodynamic model in pharmaceutical applications, is examined in terms of its performance regarding the computational pure prediction of API-polymer compatibility based on activity coefficients (API fusion properties were taken from experiments) without any binary interaction parameters fitted to API-polymer experimental data (that is, kij = 0 in all cases). This kind of prediction does not need any experimental binary information and has been underreported in the literature so far, as the routine modeling strategy used in the majority of the existing PC-SAFT applications to ASDs comprised the use of nonzero kij values. The predictive performance of PC-SAFT was systematically and thoroughly evaluated against reliable experimental data for almost 40 API-polymer combinations. We also examined the effect of different sets of PC-SAFT parameters for APIs on compatibility predictions. Quantitatively, the total average error calculated over all systems was approximately 50% in the weight fraction solubility of APIs in polymers, regardless of the specific API parametrization. The magnitude of the error for individual systems was found to vary significantly from one system to another. Interestingly, the poorest results were obtained for systems with self-associating polymers such as poly(vinyl alcohol). Such polymers can form intramolecular hydrogen bonds, which are not accounted for in the PC-SAFT variant routinely applied to ASDs (i.e., that used in this work). However, the qualitative ranking of polymers with respect to their compatibility with a given API was reasonably predicted in many cases. It was also predicted correctly that some polymers always have better compatibility with the APIs than others. Finally, possible future routes to improve the cost-performance ratio of PC-SAFT in terms of parametrization are discussed.
Collapse
Affiliation(s)
- Jáchym Pavliš
- Department of Physical Chemistry,
Faculty of Chemical Engineering, University
of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Alex Mathers
- Department of Physical Chemistry,
Faculty of Chemical Engineering, University
of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Michal Fulem
- Department of Physical Chemistry,
Faculty of Chemical Engineering, University
of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Martin Klajmon
- Department of Physical Chemistry,
Faculty of Chemical Engineering, University
of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| |
Collapse
|
13
|
Dohrn S, Kyeremateng SO, Bochmann E, Sobich E, Wahl A, Liepold B, Sadowski G, Degenhardt M. Thermodynamic Modeling of the Amorphous Solid Dispersion-Water Interfacial Layer and Its Impact on the Release Mechanism. Pharmaceutics 2023; 15:pharmaceutics15051539. [PMID: 37242781 DOI: 10.3390/pharmaceutics15051539] [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: 04/10/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
During the dissolution of amorphous solid dispersion (ASD) formulations, the gel layer that forms at the ASD/water interface strongly dictates the release of the active pharmaceutical ingredient (API) and, hence, the dissolution performance. Several studies have demonstrated that the switch of the gel layer from eroding to non-eroding behavior is API-specific and drug-load (DL)-dependent. This study systematically classifies the ASD release mechanisms and relates them to the phenomenon of the loss of release (LoR). The latter is thermodynamically explained and predicted via a modeled ternary phase diagram of API, polymer, and water, and is then used to describe the ASD/water interfacial layers (below and above the glass transition). To this end, the ternary phase behavior of the APIs, naproxen, and venetoclax with the polymer poly(vinylpyrrolidone-co-vinyl acetate) (PVPVA64) and water was modeled using the perturbed-chain statistical associating fluid theory (PC-SAFT). The glass transition was modeled using the Gordon-Taylor equation. The DL-dependent LoR was found to be caused by API crystallization or liquid-liquid phase separation (LLPS) at the ASD/water interface. If crystallization occurs, it was found that API and polymer release was impeded above a threshold DL at which the APIs crystallized directly at the ASD interface. If LLPS occurs, an API-rich phase and a polymer-rich phase are formed. Above a threshold DL, the less mobile and hydrophobic API-rich phase accumulates at the interface which prevents API release. LLPS is further influenced by the composition and glass transition temperature of the evolving phases and was investigated at 37 °C and 50 °C regarding impact of temperature of. The modeling results and LoR predictions were experimentally validated by means of dissolution experiments, microscopy, Raman spectroscopy, and size exclusion chromatography. The experimental results were found to be in very good agreement with the predicted release mechanisms deduced from the phase diagrams. Thus, this thermodynamic modeling approach represents a powerful mechanistic tool that can be applied to classify and quantitatively predict the DL-dependent LoR release mechanism of PVPVA64-based ASDs in water.
Collapse
Affiliation(s)
- Stefanie Dohrn
- AbbVie Deutschland GmbH & Co. KG, Global Pharmaceutical R&D, Knollstraße, D-67061 Ludwigshafen am Rhein, Germany
| | - Samuel O Kyeremateng
- AbbVie Deutschland GmbH & Co. KG, Global Pharmaceutical R&D, Knollstraße, D-67061 Ludwigshafen am Rhein, Germany
| | - Esther Bochmann
- AbbVie Deutschland GmbH & Co. KG, Global Pharmaceutical R&D, Knollstraße, D-67061 Ludwigshafen am Rhein, Germany
| | - Ekaterina Sobich
- AbbVie Deutschland GmbH & Co. KG, Global Pharmaceutical R&D, Knollstraße, D-67061 Ludwigshafen am Rhein, Germany
| | - Andrea Wahl
- AbbVie Deutschland GmbH & Co. KG, Global Pharmaceutical R&D, Knollstraße, D-67061 Ludwigshafen am Rhein, Germany
| | - Bernd Liepold
- AbbVie Deutschland GmbH & Co. KG, Global Pharmaceutical R&D, Knollstraße, D-67061 Ludwigshafen am Rhein, Germany
| | - Gabriele Sadowski
- Laboratory of Thermodynamics, Department of Chemical and Biochemical Engineering, TU Dortmund University, Emil-Figge-Str. 70, D-44227 Dortmund, Germany
| | - Matthias Degenhardt
- AbbVie Deutschland GmbH & Co. KG, Global Pharmaceutical R&D, Knollstraße, D-67061 Ludwigshafen am Rhein, Germany
| |
Collapse
|
14
|
Mansuri A, Münzner P, Heermant A, Patzina F, Feuerbach T, Winck J, Vermeer AWP, Hoheisel W, Böhmer R, Gainaru C, Thommes M. Molecular Dynamics and Diffusion in Amorphous Solid Dispersions Containing Imidacloprid. Mol Pharm 2023; 20:2067-2079. [PMID: 36930788 DOI: 10.1021/acs.molpharmaceut.2c01042] [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: 03/19/2023]
Abstract
The main goal of this study is to develop an experimental toolbox to estimate the self-diffusion coefficient of active ingredients (AI) in single-phase amorphous solid dispersions (ASD) close to the glass transition of the mixture using dielectric spectroscopy (DS) and oscillatory rheology. The proposed methodology is tested for a model system containing the insecticide imidacloprid (IMI) and the copolymer copovidone (PVP/VA) prepared via hot-melt extrusion. For this purpose, reorientational and the viscoelastic structural (α-)relaxation time constants of hot-melt-extruded ASDs were obtained via DS and shear rheology, respectively. These were then utilized to extract the viscosity as well as the fragility index of the dispersions as input parameters to the fractional Stokes-Einstein (F-SE) relation. Furthermore, a modified version of Almond-West (AW) formalism, originally developed to describe charge diffusion in ionic conductors, was exercised on the present model system for the estimation of the AI diffusion coefficients based on shear modulus relaxation times. Our results revealed that, at the calorimetric glass-transition temperature (Tg), the self-diffusion coefficients of the AI in the compositional range from infinite dilution up to 60 wt % IMI content lied in the narrow range of 10-18-10-20 m2 s-1, while the viscosity values of the dispersions at Tg varied between 108 Pa s and 1010 Pa s. In addition, the phase diagram of the IMI-PVP/VA system was determined using the melting point depression method via differential scanning calorimetry (DSC), while mid-infrared (IR) spectroscopy was employed to investigate the intermolecular interactions within the solid dispersions. In this respect, the findings of a modest variation in melting point at different compositions stayed in agreement with the observations of weak hydrogen bonding interactions between the AI and the polymer. Moreover, IR spectroscopy showed the intermolecular IMI-IMI hydrogen bonding to have been considerably suppressed, as a result of the spatial separation of the AI molecules within the ASDs. In summary, this study provides experimental approaches to study diffusivity in ASDs using DS and oscillatory rheology, in addition to contributing to an enhanced understanding of the interactions and phase behavior in these systems.
Collapse
Affiliation(s)
- Ali Mansuri
- Department of Biochemical and Chemical Engineering, TU Dortmund University, 44227 Dortmund, Germany.,INVITE GmbH, 51061 Cologne, Germany
| | - Philipp Münzner
- Fakultät Physik, Technische Universität Dortmund, 44221 Dortmund, Germany
| | - Anrika Heermant
- Department of Biochemical and Chemical Engineering, TU Dortmund University, 44227 Dortmund, Germany
| | - Fabian Patzina
- Department of Biochemical and Chemical Engineering, TU Dortmund University, 44227 Dortmund, Germany
| | | | - Judith Winck
- Department of Biochemical and Chemical Engineering, TU Dortmund University, 44227 Dortmund, Germany
| | | | | | - Roland Böhmer
- Fakultät Physik, Technische Universität Dortmund, 44221 Dortmund, Germany
| | - Catalin Gainaru
- Chemical Sciences Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee 37831, United States
| | - Markus Thommes
- Department of Biochemical and Chemical Engineering, TU Dortmund University, 44227 Dortmund, Germany
| |
Collapse
|
15
|
Bertoni S, Albertini B, Passerini N. Investigating the physicochemical properties of solid dispersions based on semicrystalline carriers: A case study with ketoprofen. Int J Pharm 2023; 632:122576. [PMID: 36596317 DOI: 10.1016/j.ijpharm.2022.122576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/01/2023]
Abstract
Hydrophilic semicrystalline carriers represent an alternative to amorphous polymers due to their low melting temperature, useful for the production of solid dispersions (SDs) by melting-based technologies. This research aims to compare SDs of ketoprofen (KET) and three different semicrystalline carriers (PEG, Poloxamer and Gelucire) regarding miscibility, phase behavior, molecular interactions and stability. KET was chosen owing to its low solubility and high glass forming ability. Estimation of drug-excipient miscibility was performed by Flory-Huggins theory. Negative Gibbs free energy indicated a spontaneous mixing of KET with the three carriers and miscibility in the order PEG > Poloxamer > Gelucire. SDs up to 40 % w/w of drug were produced by melting process at a temperature below KET melting point. Characterization of SDs was performed by differential scanning calorimetry, polarized light microscopy and powder X-ray diffraction. In case of PEG and Poloxamer, the drug incorporation did not affect carrier crystallinity, while KET was in the amorphous state. Differently, KET retarded the crystallization of Gelucire and at high drug loadings the SDs were amorphous and semisolid. FT-IR analysis revealed a strong interaction between KET and the three carriers. Finally, PEG-based SDs above 20 % KET loading displayed drug crystallization after 6 months of storage; while Poloxamer and Gelucire-based SDs showed KET crystallization only at 40 % KET. Due to its less hydrophilic character and limited water uptake, Gelucire showed the best stability among the three excipients.
Collapse
Affiliation(s)
- Serena Bertoni
- Department of Pharmacy and BioTechnology, PharmTech Lab, Alma Mater Studiorum-University of Bologna, Via S. Donato 19/2, 40127 Bologna, Italy
| | - Beatrice Albertini
- Department of Pharmacy and BioTechnology, PharmTech Lab, Alma Mater Studiorum-University of Bologna, Via S. Donato 19/2, 40127 Bologna, Italy
| | - Nadia Passerini
- Department of Pharmacy and BioTechnology, PharmTech Lab, Alma Mater Studiorum-University of Bologna, Via S. Donato 19/2, 40127 Bologna, Italy.
| |
Collapse
|
16
|
Chen Q, Ji Y. Thermodynamic Mechanism of Physical Stability of Amorphous Pharmaceutical Formulations. Ind Eng Chem Res 2023. [DOI: 10.1021/acs.iecr.2c02953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Qiao Chen
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing211189, People’s Republic of China
| | - Yuanhui Ji
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing211189, People’s Republic of China
| |
Collapse
|
17
|
Meiland P, Larsen BS, Knopp MM, Tho I, Rades T. A new method to determine drug-polymer solubility through enthalpy of melting and mixing. Int J Pharm 2022; 629:122391. [PMID: 36379397 DOI: 10.1016/j.ijpharm.2022.122391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/15/2022]
Abstract
In this study, a new method to determine the solubility of crystalline drugs in (amorphous) polymers is proposed. The method utilizes annealing of supersaturated amorphous solid dispersions to achieve equilibrium between dissolved and recrystallized drug. By measuring the enthalpy of melting and mixing (Hm+mix) of the recrystallized drug, the equilibrium solubility of the drug in the polymer at the annealing temperature is determined. The equilibrium solubilities at these elevated temperatures were used to extrapolate to room temperature using the Flory-Huggins model. The new Hm+mix method showed solubility predictions in line with the melting point depression (MPD) and recrystallization (RC) methods for indomethacin (IMC) -polyvinylpyrrolidone (PVP). For IMC-hydroxypropyl methylcellulose (HPMC), the MPD method plateaued rapidly, leaving only one usable data point. The RC method showed large variations in the solubility predictions possibly due to a narrow glass transition temperature (Tg) window or inaccurate Tg determination. In contrast, the new Hm+mix method showed robust solubility prediction over the entire annealing temperature range with low variation and narrow error margins after extrapolation for both drug-polymer systems. The new Hm+mix method was able to accurately determine the drug-polymer solubility of IMC-HPMC, showing promise as a new tool to determine the solubility of problematic drug-polymer systems.
Collapse
Affiliation(s)
- Peter Meiland
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| | - Bjarke Strøm Larsen
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Sem Sælands vei 3, 0371 Oslo, Norway.
| | - Matthias Manne Knopp
- Bioneer:FARMA, Department of Pharmacy, Universitetsparken 2, 2100 Copenhagen, Denmark.
| | - Ingunn Tho
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Sem Sælands vei 3, 0371 Oslo, Norway.
| | - Thomas Rades
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| |
Collapse
|
18
|
Iemtsev A, Hassouna F, Klajmon M, Mathers A, Fulem M. Compatibility of selected active pharmaceutical ingredients with poly(D, L-lactide-co-glycolide): Computational and experimental study. Eur J Pharm Biopharm 2022. [DOI: 10.1016/j.ejpb.2022.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
19
|
Klajmon M. Purely Predicting the Pharmaceutical Solubility: What to Expect from PC-SAFT and COSMO-RS? Mol Pharm 2022; 19:4212-4232. [PMID: 36136040 DOI: 10.1021/acs.molpharmaceut.2c00573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A pair of popular thermodynamic models for pharmaceutical applications, namely, the perturbed-chain statistical associating fluid theory (PC-SAFT) equation of state and the conductor-like screening model for real solvents (COSMO-RS) are thoroughly benchmarked for their performance in predicting the solubility of active pharmaceutical ingredients (APIs) in pure solvents. The ultimate goal is to provide an illustration of what to expect from these progressive frameworks when applied to the thermodynamic solubility of APIs based on activity coefficients in a purely predictive regime without specific experimental solubility data (the fusion properties of pure APIs were taken from experiments). While this kind of prediction represents the typical modus operandi of the first-principles-aided COSMO-RS, PC-SAFT is a relatively highly parametrized model that relies on experimental data, against which its pure-substance and binary interaction parameters (kij) are fitted. Therefore, to make this benchmark as fair as possible, we omitted any binary parameters of PC-SAFT (i.e., kij = 0 in all cases) and preferred pure-substance parameter sets for APIs not trained to experimental solubility data. This computational approach, together with a detailed assessment of the obtained solubility predictions against a large experimental data set, revealed that COSMO-RS convincingly outperformed PC-SAFT both qualitatively (i.e., COSMO-RS was better in solvent ranking) and quantitatively, even though the former is independent of both substance- and mixture-specific experimental data. Regarding quantitative comparison, COSMO-RS outperformed PC-SAFT for 9 of the 10 APIs and for 63% of the API-solvent systems, with root-mean-square deviations of the predicted data from the entire experimental data set being 0.82 and 1.44 log units, respectively. The results were further analyzed to expand the picture of the performance of both models with respect to the individual APIs and solvents. Interestingly, in many cases, both models were found to qualitatively incorrectly predict the direction of deviations from ideality. Furthermore, we examined how the solubility predictions from both models are sensitive to different API parametrizations.
Collapse
Affiliation(s)
- Martin Klajmon
- Department of Physical Chemistry, Faculty of Chemical Engineering, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| |
Collapse
|
20
|
Anomalous Water-Sorption Kinetics in ASDs. Pharmaceutics 2022; 14:pharmaceutics14091897. [PMID: 36145645 PMCID: PMC9505135 DOI: 10.3390/pharmaceutics14091897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 11/25/2022] Open
Abstract
Anomalous water-sorption kinetics in amorphous solid dispersions (ASDs) are caused by the slow swelling of the polymer. In this work, we used a diffusion–relaxation model with the Williams–Landel–Ferry (WLF) equation and the Arrhenius equation to predict the anomalous water-sorption kinetics in ASDs of poly(vinyl-pyrrolidone)-co-vinyl-acetate (PVPVA) and indomethacin (IND) at 25 °C. These predictions were based on the viscosities of pure PVPVA and pure IND, as well as on the water-sorption kinetics in pure PVPVA. The diffusion–relaxation model was able to predict the different types of anomalous behavior leading to a qualitative and quantitative agreement with the experimental data. Predictions and experiments indicated more pronounced anomalous two-stage water-sorption behavior in the ASDs than in pure PVPVA. This was caused by a higher viscosity of glassy ASD–water mixtures compared to glassy PVPVA–water mixtures at the same distance from their glass transition temperature. These results suggest that this ASD swells more slowly than the polymer it is composed of. The modeling approach applied in this work can be used in the future for predicting diffusion-controlled release behavior or swelling-controlled release behavior of ASDs.
Collapse
|
21
|
Predicting the Water Sorption in ASDs. Pharmaceutics 2022; 14:pharmaceutics14061181. [PMID: 35745753 PMCID: PMC9230848 DOI: 10.3390/pharmaceutics14061181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/26/2022] [Accepted: 05/30/2022] [Indexed: 02/04/2023] Open
Abstract
Water decreases the stability of amorphous solid dispersions (ASDs) and water sorption is, therefore, unwanted during ASD storage. This work suggests a methodology to predict the water-sorption isotherms and the water-sorption kinetics in amorphous pharmaceutical formulations like ASDs. We verified the validity of the proposed methodology by measuring and predicting the water-sorption curves in ASD films of polyvinylpyrrolidone-based polymers and of indomethacin. This way, the extent and the rate of water sorption in ASDs were predicted for drug loads of 0.2 and 0.5 as well as in the humidity range from 0 to 0.9 RH at 25 °C. The water-sorption isotherms and the water-sorption kinetics in the ASDs were predicted only based on the water-sorption isotherms and water-sorption kinetics in the neat polymer on the one hand and in the neat active pharmaceutical ingredient (API) on the other hand. The accurate prediction of water-sorption isotherms was ensured by combining the Perturbed-Chain Statistical Association Theory (PC-SAFT) with the Non-Equilibrium Thermodynamics of Glassy Polymers (NET-GP) approach. Water-sorption kinetics were predicted using Maxwell–Stefan diffusion coefficients of water in the ASDs.
Collapse
|
22
|
Kyeremateng SO, Voges K, Dohrn S, Sobich E, Lander U, Weber S, Gessner D, Evans RC, Degenhardt M. A Hot-Melt Extrusion Risk Assessment Classification System for Amorphous Solid Dispersion Formulation Development. Pharmaceutics 2022; 14:pharmaceutics14051044. [PMID: 35631630 PMCID: PMC9147278 DOI: 10.3390/pharmaceutics14051044] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/07/2022] [Accepted: 05/10/2022] [Indexed: 02/05/2023] Open
Abstract
Several literature publications have described the potential application of active pharmaceutical ingredient (API)–polymer phase diagrams to identify appropriate temperature ranges for processing amorphous solid dispersion (ASD) formulations via the hot-melt extrusion (HME) technique. However, systematic investigations and reliable applications of the phase diagram as a risk assessment tool for HME are non-existent. Accordingly, within AbbVie, an HME risk classification system (HCS) based on API–polymer phase diagrams has been developed as a material-sparing tool for the early risk assessment of especially high melting temperature APIs, which are typically considered unsuitable for HME. The essence of the HCS is to provide an API risk categorization framework for the development of ASDs via the HME process. The proposed classification system is based on the recognition that the manufacture of crystal-free ASD using the HME process fundamentally depends on the ability of the melt temperature to reach the API’s thermodynamic solubility temperature or above. Furthermore, we explored the API–polymer phase diagram as a simple tool for process design space selection pertaining to API or polymer thermal degradation regions and glass transition temperature-related dissolution kinetics limitations. Application of the HCS was demonstrated via HME experiments with two high melting temperature APIs, sulfamerazine and telmisartan, with the polymers Copovidone and Soluplus. Analysis of the resulting ASDs in terms of the residual crystallinity and degradation showed excellent agreement with the preassigned HCS class. Within AbbVie, the HCS concept has been successfully applied to more than 60 different APIs over the last 8 years as a robust validated risk assessment and quality-by-design (QbD) tool for the development of HME ASDs.
Collapse
|
23
|
Seiler VK, Theil F, Nagel N, van Lishaut H. Adding a new Dimension to the Amorphous Solid Dispersion Phase Diagram: Studying Dissolution Kinetics of Crystalline Drugs in a Polymer Matrix using temperature dependent XRPD and DSC. J Pharm Sci 2022; 111:2496-2504. [DOI: 10.1016/j.xphs.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 04/05/2022] [Accepted: 04/05/2022] [Indexed: 10/18/2022]
|
24
|
Borrmann D, Danzer A, Sadowski G. Measuring and Modeling Water Sorption in Amorphous Indomethacin and Ritonavir. Mol Pharm 2022; 19:998-1007. [DOI: 10.1021/acs.molpharmaceut.1c00984] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Dominik Borrmann
- Department of Chemical and Biochemical Engineering, Laboratory of Thermodynamics, TU Dortmund University, Emil-Figge-Str. 70, Dortmund D-44227, Germany
| | - Andreas Danzer
- Department of Chemical and Biochemical Engineering, Laboratory of Thermodynamics, TU Dortmund University, Emil-Figge-Str. 70, Dortmund D-44227, Germany
| | - Gabriele Sadowski
- Department of Chemical and Biochemical Engineering, Laboratory of Thermodynamics, TU Dortmund University, Emil-Figge-Str. 70, Dortmund D-44227, Germany
| |
Collapse
|
25
|
Factors Influencing the Crystallization-Onset Time of Metastable ASDs. Pharmaceutics 2022; 14:pharmaceutics14020269. [PMID: 35214002 PMCID: PMC8879851 DOI: 10.3390/pharmaceutics14020269] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 02/04/2023] Open
Abstract
In formulation development, amorphous solid dispersions (ASD) are considered to improve the bioavailability of poorly water-soluble active pharmaceutical ingredients (APIs). However, the crystallization of APIs often limits long-term stability and thus the shelf life of ASDs. It has already been shown earlier that the long-term stability of ASDs strongly depends on the storage conditions (relative humidity, temperature), the manufacturing methods, and the resulting particle sizes. In this work, ASDs composed of the model APIs Griseofulvin (GRI) or Itraconazole (ITR) and the polymers poly (vinylpyrrolidone-co-vinyl acetate) (PVPVA) or Soluplus® were manufactured via spray drying and hot-melt extrusion. Each API/polymer combination was manufactured using the two manufacturing methods with at least two different API loads and two particle-size distributions. It was a priori known that these ASDs were metastable and would crystallize over time, even in the dry stage. The amount of water absorbed by the ASD from humid air (40 °C/75% relative humidity), the solubility of the API in the ASD at humid conditions, and the resulting glass-transition temperature were predicted using the Perturbed-Chain Statistical Associating Fluid Theory (PC-SAFT) and the Gordon–Taylor approach, respectively. The onset of crystallization was determined via periodic powder X-ray diffraction (PXRD) measurements. It was shown that simple heuristics such as “larger particles always crystallize later than smaller particles” are correct within one manufacturing method but cannot be transferred from one manufacturing method to another. Moreover, amorphous phase separation in the ASDs was shown to also influence their crystallization kinetics. Counterintuitively, phase separation accelerated the crystallization time, which could be explained by the glass-transition temperatures of the evolving phases.
Collapse
|
26
|
Ball milling and hot-melt extrusion of indomethacin-L-arginine-vinylpyrrolidone-vinyl acetate copolymer: Solid-state properties and dissolution performance. Int J Pharm 2021; 613:121424. [PMID: 34968683 DOI: 10.1016/j.ijpharm.2021.121424] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 11/23/2022]
Abstract
Commonly applied approaches to enhance the dissolution properties of low water-soluble crystalline active pharmaceutical ingredients (APIs) include their amorphization by incorporation into a polymeric matrix and the formation of amorphous solid dispersions (ASDs), or blending APIs with low-molecular-weight excipients and the formation of a co-amorphous system. This study focused on the preparation and characterization of binary (consisting of indomethacin (IND) and polymer - copovidone (PVP VA 64), as a carrier, or amino acid - L-arginine (ARG), as a co-former) and ternary (comprising the same API, polymer, and amino acid) formulations. Formulations were produced by ball milling (BM) and/or hot-melt extrusion (HME), and extensive physicochemical characterization was performed. Specifically, the physicochemical and solid-state properties of a model IND-ARG system incorporated into a polymeric matrix of PVP VA 64 by HME and BM as well as by combined BM/HME method together with the impact of the preparation strategy on the dissolution profiles and long-term physical stability were investigated. Ball-milled binary and ternary formulations were found to be amorphous. The residual crystals corresponding to IND-ARG salt were identified in the ternary formulations produced via HME. Despite the presence of a crystalline phase, dissolution tests showed that ternary systems prepared by HME exhibited improved IND solubility when compared to pure crystalline IND and their corresponding physical mixture. None of the binary and ternary formulations that were initially fully amorphous did undergo recrystallization during the entire period of preservation (minimum of 12 months) in dry conditions at 25℃.
Collapse
|
27
|
Sadeghi M, Cascella F, Tenberg V, Seidel-Morgenstern A, Lorenz H. Solubility analysis of pharmaceuticals guaifenesin, ketoprofen, and artemisinin in different solvents. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.117503] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
28
|
Influence of excipients on thermodynamic phase behavior of pharmaceutical/solvent systems: Molecular thermodynamic model prediction. Chem Eng Sci 2021. [DOI: 10.1016/j.ces.2021.116798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
29
|
Ji Y, Hao D, Luebbert C, Sadowski G. Insights into influence mechanism of polymeric excipients on dissolution of drug formulations: A molecular interaction‐based theoretical model analysis and prediction. AIChE J 2021. [DOI: 10.1002/aic.17372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Yuanhui Ji
- Jiangsu Province Hi‐Tech Key Laboratory for Bio‐medical Research, School of Chemistry and Chemical Engineering, Southeast University Nanjing China
| | - Dule Hao
- Jiangsu Province Hi‐Tech Key Laboratory for Bio‐medical Research, School of Chemistry and Chemical Engineering, Southeast University Nanjing China
| | - Christian Luebbert
- TU Dortmund, Department of Biochemical and Chemical Engineering Laboratory of Thermodynamics Dortmund Germany
| | - Gabriele Sadowski
- TU Dortmund, Department of Biochemical and Chemical Engineering Laboratory of Thermodynamics Dortmund Germany
| |
Collapse
|
30
|
Ge K, Ji Y. Novel Computational Approach by Combining Machine Learning with Molecular Thermodynamics for Predicting Drug Solubility in Solvents. Ind Eng Chem Res 2021. [DOI: 10.1021/acs.iecr.1c00998] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Kai Ge
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, People’s Republic of China
| | - Yuanhui Ji
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, People’s Republic of China
| |
Collapse
|
31
|
Thakore SD, Akhtar J, Jain R, Paudel A, Bansal AK. Analytical and Computational Methods for the Determination of Drug-Polymer Solubility and Miscibility. Mol Pharm 2021; 18:2835-2866. [PMID: 34041914 DOI: 10.1021/acs.molpharmaceut.1c00141] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In the pharmaceutical industry, poorly water-soluble drugs require enabling technologies to increase apparent solubility in the biological environment. Amorphous solid dispersion (ASD) has emerged as an attractive strategy that has been used to market more than 20 oral pharmaceutical products. The amorphous form is inherently unstable and exhibits phase separation and crystallization during shelf life storage. Polymers stabilize the amorphous drug by antiplasticization, reducing molecular mobility, reducing chemical potential of drug, and increasing glass transition temperature in ASD. Here, drug-polymer miscibility is an important contributor to the physical stability of ASDs. The current Review discusses the basics of drug-polymer interactions with the major focus on the methods for the evaluation of solubility and miscibility of the drug in the polymer. Methods for the evaluation of drug-polymer solubility and miscibility have been classified as thermal, spectroscopic, microscopic, solid-liquid equilibrium-based, rheological, and computational methods. Thermal methods have been commonly used to determine the solubility of the drug in the polymer, while other methods provide qualitative information about drug-polymer miscibility. Despite advancements, the majority of these methods are still inadequate to provide the value of drug-polymer miscibility at room temperature. There is still a need for methods that can accurately determine drug-polymer miscibility at pharmaceutically relevant temperatures.
Collapse
Affiliation(s)
- Samarth D Thakore
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Junia Akhtar
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Ranjna Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Amrit Paudel
- Research Center Pharmaceutical Engineering (RCPE) GmbH, Inffeldgasse 13, 8010 Graz, Austria.,Institute for Process and Particle Engineering, Graz University of Technology, Inffeldgasse 13, 8010 Graz, Austria
| | - Arvind K Bansal
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| |
Collapse
|
32
|
Dohrn S, Rawal P, Luebbert C, Lehmkemper K, Kyeremateng SO, Degenhardt M, Sadowski G. Predicting process design spaces for spray drying amorphous solid dispersions. Int J Pharm X 2021; 3:100072. [PMID: 33855291 PMCID: PMC8027772 DOI: 10.1016/j.ijpx.2021.100072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 02/06/2021] [Indexed: 11/01/2022] Open
Abstract
Amorphous solid dispersions (ASDs) are commonly manufactured using spray-drying processes. The product quality can be decisively influenced by the choice of process parameters. Following the quality-by-design approach, the identification of the spray-drying process design space is thus an integral task in drug product development. Aiming a solvent-free and homogeneous ASD, API crystallization and amorphous phase separation needs to be avoided during drying. This publication provides a predictive approach for determining spray-drying process conditions via considering thermodynamic driving forces for solvent drying as well as ASD-specific API/polymer/solvent interactions and glass transitions. The ternary API/polymer/solvent phase behavior was calculated using the Perturbed-Chain Statistical Associating Theory (PC-SAFT) and combined with mass and energy balances to find appropriate spray-drying conditions. A process design space was identified for the ASDs of ritonavir and naproxen with either poly(vinylpyrrolidone) or poly(vinylpyrrolidone-co-vinylacetate) spray dried from the solvents acetone, dichloromethane, or ethanol.
Collapse
Affiliation(s)
- Stefanie Dohrn
- TU Dortmund University, Department of Biochemical and Chemical Engineering, Laboratory of Thermodynamics, Emil-Figge-Str. 70, D-44227 Dortmund, Germany
| | - Pranay Rawal
- TU Dortmund University, Department of Biochemical and Chemical Engineering, Laboratory of Thermodynamics, Emil-Figge-Str. 70, D-44227 Dortmund, Germany
| | - Christian Luebbert
- TU Dortmund University, Department of Biochemical and Chemical Engineering, Laboratory of Thermodynamics, Emil-Figge-Str. 70, D-44227 Dortmund, Germany
| | - Kristin Lehmkemper
- AbbVie Deutschland GmbH & Co. KG, Global Pharmaceutical R&D, Knollstraße, D-67061 Ludwigshafen am Rhein, Germany
| | - Samuel O Kyeremateng
- AbbVie Deutschland GmbH & Co. KG, Global Pharmaceutical R&D, Knollstraße, D-67061 Ludwigshafen am Rhein, Germany
| | - Matthias Degenhardt
- AbbVie Deutschland GmbH & Co. KG, Global Pharmaceutical R&D, Knollstraße, D-67061 Ludwigshafen am Rhein, Germany
| | - Gabriele Sadowski
- TU Dortmund University, Department of Biochemical and Chemical Engineering, Laboratory of Thermodynamics, Emil-Figge-Str. 70, D-44227 Dortmund, Germany
| |
Collapse
|
33
|
Mathers A, Hassouna F, Klajmon M, Fulem M. Comparative Study of DSC-Based Protocols for API-Polymer Solubility Determination. Mol Pharm 2021; 18:1742-1757. [PMID: 33656884 DOI: 10.1021/acs.molpharmaceut.0c01232] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Knowledge of the active pharmaceutical ingredient (API) solubility in a polymer is imperative for successful amorphous solid dispersion design and formulation but acquiring this information at storage temperature is challenging. Various solubility determination methods have been established, which utilize differential scanning calorimetry (DSC). In this work, three commonly used DSC-based protocols [i.e., melting point depression (MPD), recrystallization, and zero-enthalpy extrapolation (Z-EE)] and a method that we have developed called "step-wise dissolution" (S-WD) were analyzed. For temperature-composition phase diagram construction, two glass-transition temperature equations (i.e., those of Gordon-Taylor and Kwei) and three solid-liquid equilibrium curve modeling approaches [i.e., the Flory-Huggins model, an empirical equation, and the perturbed-chain statistical associating fluid theory (PC-SAFT) equation of state (EOS)] were considered. Indomethacin (IND) and Kollidon 12 PF (PVP K12) were selected as the API and polymer, respectively. An annealing time investigation revealed that the IND-PVP K12 dissolution process was remarkably faster than demixing, which contradicted previously published statements. Thus, the recrystallization method overestimated the solubility of IND in PVP K12 when a 2-h time of annealing was set as the benchmark. Likewise, the MPD and Z-EE methods overestimated the API solubility because of unreliable IND melting endotherm evaluation at lower API loadings and a relatively slow heating rate, respectively. When the experimental results obtained using the S-WD method (in conjunction with the Kwei equation) were applied to the PC-SAFT EOS, which was regarded as the most reliable combination, the predicted IND solubility in PVP K12 at T = 25 °C was approximately 40 wt %. When applicable, the S-WD method offers the advantage of using a limited number of DSC sample pans and API-polymer physical mixture compositions, which is both cost- and time-effective.
Collapse
Affiliation(s)
- Alex Mathers
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Fatima Hassouna
- Faculty of Chemical Engineering, University of Chemistry and Technology, Prague, Technická 3, 166 28 Prague 6, Czech Republic
| | - Martin Klajmon
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Michal Fulem
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| |
Collapse
|
34
|
Ge K, Ji Y, Lu X. A novel interfacial thermodynamic model for predicting solubility of nanoparticles coated by stabilizers. Chin J Chem Eng 2021. [DOI: 10.1016/j.cjche.2020.10.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
35
|
Butreddy A, Bandari S, Repka MA. Quality-by-design in hot melt extrusion based amorphous solid dispersions: An industrial perspective on product development. Eur J Pharm Sci 2021; 158:105655. [PMID: 33253883 PMCID: PMC7855693 DOI: 10.1016/j.ejps.2020.105655] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/21/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023]
Abstract
An industrially feasible approach to overcome the solubility and bioavailability limitations of poorly soluble active pharmaceutical ingredients is the development of amorphous solid dispersions (ASDs) using hot-melt extrusion (HME) technique. The application of Quality by Design (QbD) had a profound impact on the development of HME-based ASDs. The formulation and process optimization of ASDs manufactured via HME techniques require an understanding of critical quality attributes, critical material attributes, critical process parameters, risk assessment tools, and experimental designs. The knowledge gained from each of these QbD elements helps ensure the consistency of product quality. The selection and implementation of appropriate Design of Experiments (DoE) methodology to screen and optimize the formulation and process variables remain a major challenge. This review provides a comprehensive overview on QbD concepts in HME-based ASDs with an emphasis on DoE methodologies. Further, the information provided in this review can assist researchers in selecting a suitable design with optimal experimental conditions. Specifically, this review has focused on the prediction of drug-polymer miscibility, the elements and sequence of QbD, and various screening and optimization designs, to provide insights into the formulation and process variables that are encountered routinely in the production of HME-based ASDs.
Collapse
Affiliation(s)
- Arun Butreddy
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38677, USA
| | - Suresh Bandari
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38677, USA
| | - Michael A Repka
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38677, USA; Pii Center for Pharmaceutical Technology, The University of Mississippi, University, MS 38677, USA.
| |
Collapse
|
36
|
Castillo-Henríquez L, Castro-Alpízar J, Lopretti-Correa M, Vega-Baudrit J. Exploration of Bioengineered Scaffolds Composed of Thermo-Responsive Polymers for Drug Delivery in Wound Healing. Int J Mol Sci 2021; 22:1408. [PMID: 33573351 PMCID: PMC7866792 DOI: 10.3390/ijms22031408] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/13/2021] [Accepted: 01/27/2021] [Indexed: 02/06/2023] Open
Abstract
Innate and adaptive immune responses lead to wound healing by regulating a complex series of events promoting cellular cross-talk. An inflammatory response is presented with its characteristic clinical symptoms: heat, pain, redness, and swelling. Some smart thermo-responsive polymers like chitosan, polyvinylpyrrolidone, alginate, and poly(ε-caprolactone) can be used to create biocompatible and biodegradable scaffolds. These processed thermo-responsive biomaterials possess 3D architectures similar to human structures, providing physical support for cell growth and tissue regeneration. Furthermore, these structures are used as novel drug delivery systems. Locally heated tumors above the polymer lower the critical solution temperature and can induce its conversion into a hydrophobic form by an entropy-driven process, enhancing drug release. When the thermal stimulus is gone, drug release is reduced due to the swelling of the material. As a result, these systems can contribute to the wound healing process in accelerating tissue healing, avoiding large scar tissue, regulating the inflammatory response, and protecting from bacterial infections. This paper integrates the relevant reported contributions of bioengineered scaffolds composed of smart thermo-responsive polymers for drug delivery applications in wound healing. Therefore, we present a comprehensive review that aims to demonstrate these systems' capacity to provide spatially and temporally controlled release strategies for one or more drugs used in wound healing. In this sense, the novel manufacturing techniques of 3D printing and electrospinning are explored for the tuning of their physicochemical properties to adjust therapies according to patient convenience and reduce drug toxicity and side effects.
Collapse
Affiliation(s)
- Luis Castillo-Henríquez
- National Laboratory of Nanotechnology (LANOTEC), National Center for High Technology (CeNAT), 1174-1200 San José, Costa Rica;
- Physical Chemistry Laboratory, Faculty of Pharmacy, University of Costa Rica, 11501-2060 San José, Costa Rica
| | - Jose Castro-Alpízar
- Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Costa Rica, 11501-2060 San José, Costa Rica;
| | - Mary Lopretti-Correa
- Nuclear Research Center, Faculty of Science, Universidad de la República (UdelaR), 11300 Montevideo, Uruguay;
| | - José Vega-Baudrit
- National Laboratory of Nanotechnology (LANOTEC), National Center for High Technology (CeNAT), 1174-1200 San José, Costa Rica;
- Laboratory of Polymers (POLIUNA), Chemistry School, National University of Costa Rica, 86-3000 Heredia, Costa Rica
| |
Collapse
|
37
|
Schönfeld B, Westedt U, Wagner KG. Vacuum drum drying - A novel solvent-evaporation based technology to manufacture amorphous solid dispersions in comparison to spray drying and hot melt extrusion. Int J Pharm 2021; 596:120233. [PMID: 33484914 DOI: 10.1016/j.ijpharm.2021.120233] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 10/22/2022]
Abstract
In this study, a novel solvent-evaporation based technology to manufacture amorphous solid dispersions (ASDs) called vacuum drum drying (VDD) was assessed in comparison to the conventional technologies hot-melt extrusion (HME) and spray drying (SD). Ritonavir (15%w/w) embedded in copovidone/sorbitan monolaurate was used to investigate the impact on the ASD quality, material properties and in-vitro dissolution. All ASDs met the critical quality criteria: absence of drug substance related crystallinity, residual solvents below ICH limit (SD, VDD) and degradation products within specification limits. Clear differences in material properties such as particle morphology and size distribution, powder densities and flowability properties were observed. Overall, the milled extrudate showed superior material properties in terms of downstream processability. The VDD intermediate performed slightly better in terms of flowability and electrostatic behavior compared to the spray dried while showing comparably unfavorable densities. However, the dissolution data suggested no significant difference between the ASDs prepared by HME, SD, and VDD and thus, no change in bioavailability is expected. In conclusion, the VDD technology might be a viable alternative to manufacture ASDs - especially for thermosensitive and shear-sensitive compounds with potential to process formulations with high solid loads and viscosities while exhibiting higher throughputs at a lower footprint.
Collapse
Affiliation(s)
- Barbara Schönfeld
- Department of Pharmaceutical Technology, University of Bonn, Gerhard-Domagk-Straße 3, 53121 Bonn, Germany; AbbVie Deutschland GmbH & Co. KG, Knollstrasse 50, 67061 Ludwigshafen, Germany.
| | - Ulrich Westedt
- AbbVie Deutschland GmbH & Co. KG, Knollstrasse 50, 67061 Ludwigshafen, Germany.
| | - Karl G Wagner
- Department of Pharmaceutical Technology, University of Bonn, Gerhard-Domagk-Straße 3, 53121 Bonn, Germany.
| |
Collapse
|
38
|
Luebbert C, Stoyanov E, Sadowski G. Phase behavior of ASDs based on hydroxypropyl cellulose. Int J Pharm X 2021; 3:100070. [PMID: 33409486 PMCID: PMC7773875 DOI: 10.1016/j.ijpx.2020.100070] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/02/2022] Open
Abstract
Novel polymeric carriers for amorphous solid dispersions (ASDs) are highly demanded in pharmaceutical industry to improve the bioavailability of poorly-soluble drug candidates. Besides established polymer candidates, hydroxypropyl celluloses (HPC) comes more and more into the focus of ASD production since they have the availability to stabilize drug molecules in aqueous media against crystallization. The thermodynamic long-term stability of HPC ASDs with itraconazole and fenofibrate was predicted in this work with PC-SAFT and compared to three-months enduring long-term stability studies. The glass-transition temperature is a crucial attribute of a polymer, but in case of HPC hardly detectable by differential scanning calorimetry. By investigating the glass transition of HPC blends with a miscible polymer, we were for the first time able to estimate the HPC glass transition. Although both, fenofibrate and itraconazole reveal a very low crystalline solubility in HPC regardless of the HPC molecular weight, we observed that low-molecular weight HPC grades such as HPC-UL prevent fenofibrate crystallization for a longer period than the higher molecular weight HPC grades. As predicted, the ASDs with higher drug load underwent amorphous phase separation according to the differential scanning calorimetry thermograms. This work thus showed that it is possible to predict critical drug loads above which amorphous phase separation and/or crystallization occurs in HPC ASDs.
Collapse
Affiliation(s)
| | - Edmont Stoyanov
- Nisso Chemical Europe GmbH, Berliner Allee 42, D-40212 Düsseldorf, Germany
| | - Gabriele Sadowski
- amofor GmbH, Otto-Hahn-Str. 15, D-44227 Dortmund, Germany.,TU Dortmund University, Laboratory of Thermodynamics, Emil-Figge-Str. 70, D-44227 Dortmund, Germany
| |
Collapse
|
39
|
Veith H, Wiechert F, Luebbert C, Sadowski G. Combining crystalline and polymeric excipients in API solid dispersions - Opportunity or risk? Eur J Pharm Biopharm 2020; 158:323-335. [PMID: 33296719 DOI: 10.1016/j.ejpb.2020.11.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/23/2020] [Accepted: 11/28/2020] [Indexed: 11/29/2022]
Abstract
Amorphous solid dispersions (ASDs) are often metastable against crystallization of the active pharmaceutical ingredient (API) and thus might undergo unwanted changes during storage. The crystallization tendency of ASDs is influenced by the API crystallization driving force (CDF) and the mobility of the molecules in the ASD. Low molecular weight-excipients are known to stabilize amorphous APIs in so-called co-amorphous formulations. Due to their success in stabilizing co-amorphous APIs, low-molecular weight excipients might also enhance the stability of polymeric ASDs. In this work, we investigated the potential of combined low-molecular weight excipient/polymer formulations with in-silico tools and validated the predictions with long-term stability tests of the most promising excipient/polymer combinations. The considered critical quality attributes for the ASDs were the occurrence of amorphous phase separation, API CDF, and molecular mobility in the ASD. As an example, carbamazepine/polyvinylpyrrolidone ASDs were investigated combined with the excipients fructose, lactose, sucrose, trehalose, saccharin, tryptophan, and urea. Although all excipients had a negative impact on the ASD stability, saccharin still turned out to be the most promising one. Long-term stability studies with ASDs containing either saccharin or tryptophan verified -in agreement to the predictions- that API crystallization occurred faster than in the reference ASDs without additional excipient. This work showed that the addition of crystalline excipients to polymeric ASDs might not only offer opportunities but might also bear risks for the long-term stability of the ASD, even though the crystalline excipient stabilizes the polymer-free API. Consequently, excipients should be evaluated based on the thermodynamic phase behavior of the individual mixture of API/polymer/excipient, rather than based on pure-component properties of the excipient only. In-silico predictions proposed in this work remarkably decrease the number of screening tests for identifying suitable formulation excipients.
Collapse
Affiliation(s)
- Heiner Veith
- TU Dortmund University, Department of Chemical and Biochemical Engineering, Laboratory of Thermodynamics, Emil-Figge-Str. 70, D-44227 Dortmund, Germany
| | - Felix Wiechert
- TU Dortmund University, Department of Chemical and Biochemical Engineering, Laboratory of Thermodynamics, Emil-Figge-Str. 70, D-44227 Dortmund, Germany
| | - Christian Luebbert
- TU Dortmund University, Department of Chemical and Biochemical Engineering, Laboratory of Thermodynamics, Emil-Figge-Str. 70, D-44227 Dortmund, Germany
| | - Gabriele Sadowski
- TU Dortmund University, Department of Chemical and Biochemical Engineering, Laboratory of Thermodynamics, Emil-Figge-Str. 70, D-44227 Dortmund, Germany.
| |
Collapse
|
40
|
Tang S, Ji Y, Ge K. Crystallization Kinetics and Mechanisms of Calcium Sulfate Dihydrate: Experimental Investigation and Theoretical Analysis. Ind Eng Chem Res 2020. [DOI: 10.1021/acs.iecr.0c04220] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Shuai Tang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, People’s Republic of China
| | - Yuanhui Ji
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, People’s Republic of China
| | - Kai Ge
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, People’s Republic of China
| |
Collapse
|
41
|
Brinkmann J, Exner L, Luebbert C, Sadowski G. In-Silico Screening of Lipid-Based Drug Delivery Systems. Pharm Res 2020; 37:249. [PMID: 33230602 PMCID: PMC7683453 DOI: 10.1007/s11095-020-02955-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/09/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE This work proposes an in-silico screening method for identifying promising formulation candidates in complex lipid-based drug delivery systems (LBDDS). METHOD The approach is based on a minimum amount of experimental data for API solubilites in single excipients. Intermolecular interactions between APIs and excipients as well as between different excipients were accounted for by the Perturbed-Chain Statistical Associating Fluid Theory. The approach was applied to the in-silico screening of lipid-based formulations for ten model APIs (fenofibrate, ibuprofen, praziquantel, carbamazepine, cinnarizine, felodipine, naproxen, indomethacin, griseofulvin and glibenclamide) in mixtures of up to three out of nine excipients (tricaprylin, Capmul MCM, caprylic acid, Capryol™ 90, Lauroglycol™ FCC, Kolliphor TPGS, polyethylene glycol, carbitol and ethanol). RESULTS For eight out of the ten investigated model APIs, the solubilities in the final formulations could be enhanced by up to 100 times compared to the solubility in pure tricaprylin. Fenofibrate, ibuprofen, praziquantel, carbamazepine are recommended as type I formulations, whereas cinnarizine and felodipine showed a distinctive solubility gain in type II formulations. Increased solubility was found for naproxen and indomethacin in type IIIb and type IV formulations. The solubility of griseofulvin and glibenclamide could be slightly enhanced in type IIIb formulations. The experimental validation agreed very well with the screening results. CONCLUSION The API solubility individually depends on the choice of excipients. The proposed in-silico-screening approach allows formulators to quickly determine most-appropriate types of lipid-based formulations for a given API with low experimental effort. Graphical abstract.
Collapse
Affiliation(s)
- Joscha Brinkmann
- TU Dortmund University, Laboratory of Thermodynamics, Emil-Figge-Str. 70, D-44227, Dortmund, Germany
| | - Lara Exner
- TU Dortmund University, Laboratory of Thermodynamics, Emil-Figge-Str. 70, D-44227, Dortmund, Germany
| | - Christian Luebbert
- TU Dortmund University, Laboratory of Thermodynamics, Emil-Figge-Str. 70, D-44227, Dortmund, Germany
| | - Gabriele Sadowski
- TU Dortmund University, Laboratory of Thermodynamics, Emil-Figge-Str. 70, D-44227, Dortmund, Germany.
| |
Collapse
|
42
|
Dohrn S, Luebbert C, Lehmkemper K, Kyeremateng SO, Degenhardt M, Sadowski G. Solvent influence on the phase behavior and glass transition of Amorphous Solid Dispersions. Eur J Pharm Biopharm 2020; 158:132-142. [PMID: 33212185 DOI: 10.1016/j.ejpb.2020.11.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/06/2020] [Indexed: 11/28/2022]
Abstract
Understanding the long-term stability of amorphous solid dispersions (ASDs) is important for their successful approval for market. ASD stability does not only depend on the interplay between the active pharmaceutical ingredient (API) and the polymer in the final formulation but may already be disadvantageously influenced by process steps during the production (e.g. selection of inappropriate solvent for spray drying). Residual solvent can affect the API solubility in the polymer, molecular mobility (by influencing the glass-transition temperature) and induce liquid-liquid phase separation. Enhanced mobility in the ASD due to residual solvent can promote recrystallization in ASDs. The removal of residual solvent can be expensive, time-consuming, and usually requires secondary drying procedures to fulfil the regulatory requirements. The aim of this work is to predict the API solubility in polymer-solvent mixtures, solvent influence on the glass transition, and the occurrence of liquid-liquid phase separation of solvent-loaded ASDs using the thermodynamic model PC-SAFT and to experimentally validate these predictions. ASDs containing the APIs ritonavir or naproxen and the polymers poly(vinylpyrrolidone), poly (vinylpyrrolidone-co-vinyl acetate), or hydroxypropyl methylcellulose acetate succinate were spray-dried using the solvents acetone, ethanol, and dichloromethane. API solubility, sorption behavior, liquid-liquid phase separation and glass transition in the ternary API/polymer/solvent mixtures were predicted based on the binary phase behavior between API/solvent, API/polymer, and polymer/solvent and successfully validated experimentally using dynamic vapor sorption (DVS), and Raman spectroscopy. Thus, the presented methodology allows for an in-silico selection of appropriate solvent systems for solvent-based ASD preparation based on a limited amount of experimental data for binary systems only.
Collapse
Affiliation(s)
- Stefanie Dohrn
- Department of Biochemical and Chemical Engineering, Laboratory of Thermodynamics, TU Dortmund University, Emil-Figge-Str. 70, D-44227 Dortmund, Germany
| | - Christian Luebbert
- Department of Biochemical and Chemical Engineering, Laboratory of Thermodynamics, TU Dortmund University, Emil-Figge-Str. 70, D-44227 Dortmund, Germany
| | - Kristin Lehmkemper
- AbbVie Deutschland GmbH & Co. KG, Global Pharmaceutical R&D, Knollstraße, D-67061 Ludwigshafen am Rhein, Germany
| | - Samuel O Kyeremateng
- AbbVie Deutschland GmbH & Co. KG, Global Pharmaceutical R&D, Knollstraße, D-67061 Ludwigshafen am Rhein, Germany.
| | - Matthias Degenhardt
- AbbVie Deutschland GmbH & Co. KG, Global Pharmaceutical R&D, Knollstraße, D-67061 Ludwigshafen am Rhein, Germany
| | - Gabriele Sadowski
- Department of Biochemical and Chemical Engineering, Laboratory of Thermodynamics, TU Dortmund University, Emil-Figge-Str. 70, D-44227 Dortmund, Germany.
| |
Collapse
|
43
|
Iemtsev A, Hassouna F, Mathers A, Klajmon M, Dendisová M, Malinová L, Školáková T, Fulem M. Physical stability of hydroxypropyl methylcellulose-based amorphous solid dispersions: Experimental and computational study. Int J Pharm 2020; 589:119845. [DOI: 10.1016/j.ijpharm.2020.119845] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 01/26/2023]
|
44
|
Li N, Cape JL, Mankani BR, Zemlyanov DY, Shepard KB, Morgen MM, Taylor LS. Water-Induced Phase Separation of Spray-Dried Amorphous Solid Dispersions. Mol Pharm 2020; 17:4004-4017. [PMID: 32931293 PMCID: PMC7539301 DOI: 10.1021/acs.molpharmaceut.0c00798] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
![]()
Spray
drying is widely used in the manufacturing of amorphous solid
dispersion (ASD) systems due to its fast drying rate, enabling kinetic
trapping of the drug in amorphous form. Spray-drying conditions, such
as solvent composition, can have a profound impact on the properties
of spray-dried dispersions. In this study, the phase behavior of spray-dried
dispersions from methanol and methanol–water mixtures was assessed
using ritonavir and copovidone [poly(vinylpyrrolidone-co-vinyl acetate)
(PVPVA)] as dispersion components. The resultant ASDs were characterized
using differential scanning calorimetry (DSC), fluorescence spectroscopy,
X-ray photoelectron spectroscopy (XPS), as well as surface-normalized
dissolution rate (SNDR) measurements. Quaternary phase diagrams were
calculated using a four-component Flory–Huggins model. It was
found that the addition of water to the solvent system can lead to
phase separation during the spray-drying process. A 10:90 H2O/MeOH solvent system caused a minor extent of phase separation.
Phase heterogeneity in the 50 and 75% drug loading ASDs prepared from
this spray solvent can be detected using DSC but not with other techniques
used. The 25% drug loading system did not show phase heterogeneity
in solid-state characterization but exhibited a compromised dissolution
rate compared to that of the miscible ASD prepared from H2O-free solvent. This is possibly due to the formation of slow-releasing
drug-rich phases upon phase separation. ASDs prepared with a 60:40
H2O/MeOH solvent mixture showed phase heterogeneity with
all analytical methods used. The surface composition of dispersion
particles as measured by fluorescence spectroscopy and XPS showed
good agreement, suggesting surface drug enrichment of the spray-dried
ASD particles prepared from this solvent system. Calculated phase
diagrams and drying trajectories were consistent with experimental
observations, suggesting that small variations in solvent composition
may cause significant changes in ASD phase behavior during drying.
These findings should aid in spray-drying process development for
ASD manufacturing and can be applied broadly to assess the risk of
phase separation for spray-drying systems using mixed organic solvents
or other solvent-based processes.
Collapse
Affiliation(s)
- Na Li
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States.,Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road Unit 3092, Storrs, Connecticut 06269, United States
| | - Jonathan L Cape
- Research & Development, Lonza Pharma and Biotech, 1201 NW Wall Street, Suite 200, Bend, Oregon 97703, United States
| | - Bharat R Mankani
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States.,MarqMetrix Inc., Emerald Landing, 2157 N Northlake Way #240, Seattle, Washington 98103, United States
| | - Dmitry Y Zemlyanov
- Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana 47907, United States
| | - Kimberly B Shepard
- Research & Development, Lonza Pharma and Biotech, 1201 NW Wall Street, Suite 200, Bend, Oregon 97703, United States
| | - Michael M Morgen
- Research & Development, Lonza Pharma and Biotech, 1201 NW Wall Street, Suite 200, Bend, Oregon 97703, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|
45
|
Castillo-Henríquez L, Vargas-Zúñiga R, Pacheco-Molina J, Vega-Baudrit J. Electrospun nanofibers: A nanotechnological approach for drug delivery and dissolution optimization in poorly water-soluble drugs. ADMET AND DMPK 2020; 8:325-353. [PMID: 35300196 PMCID: PMC8915594 DOI: 10.5599/admet.844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/02/2020] [Indexed: 01/02/2023] Open
Abstract
Electrospinning is a novel and sophisticated technique for the production of nanofibers with high surface area, extreme porous structure, small pore size, and surface morphologies that make them suitable for biomedical and bioengineering applications, which can provide solutions to current drug delivery issues of poorly water-soluble drugs. Electrospun nanofibers can be obtained through different methods asides from the conventional one, such as coaxial, multi-jet, side by side, emulsion, and melt electrospinning. In general, the application of an electric potential to a polymer solution causes a charged liquid jet that moves downfield to an oppositely charged collector, where the nanofibers are deposited. Plenty of polymers that differ in their origin, degradation character and water affinity are used during the process. Physicochemical properties of the drug, polymer(s), and solvent systems need to be addressed to guarantee successful manufacturing. Therefore, this review summarizes the recent progress in electrospun nanofibers for their use as a nanotechnological tool for dissolution optimization and drug delivery systems for poorly water-soluble drugs.
Collapse
Affiliation(s)
- Luis Castillo-Henríquez
- Physical Chemistry Laboratory, Faculty of Pharmacy, University of Costa Rica, 11501-2060, San José, Costa Rica
- National Laboratory of Nanotechnology (LANOTEC), National Center for High Technology (CeNAT), 1174-1200, San José, Costa Rica
| | - Rolando Vargas-Zúñiga
- Physical Chemistry Laboratory, Faculty of Pharmacy, University of Costa Rica, 11501-2060, San José, Costa Rica
| | - Jorge Pacheco-Molina
- Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Costa Rica, 11501-2060, San José, Costa Rica
| | - Jose Vega-Baudrit
- National Laboratory of Nanotechnology (LANOTEC), National Center for High Technology (CeNAT), 1174-1200, San José, Costa Rica
- Laboratory of Polymers (POLIUNA), Chemistry School, National University of Costa Rica, 86-3000, Heredia, Costa Rica
| |
Collapse
|
46
|
Ge K, Ji Y, Tang S. Crystallization Kinetics and Mechanism of Magnesium Ammonium Phosphate Hexahydrate: Experimental Investigation and Chemical Potential Gradient Model Analysis and Prediction. Ind Eng Chem Res 2020. [DOI: 10.1021/acs.iecr.0c01840] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Kai Ge
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, People’s Republic of China
| | - Yuanhui Ji
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, People’s Republic of China
| | - Shuai Tang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, People’s Republic of China
| |
Collapse
|
47
|
Matić J, Paudel A, Bauer H, Garcia RAL, Biedrzycka K, Khinast JG. Developing HME-Based Drug Products Using Emerging Science: a Fast-Track Roadmap from Concept to Clinical Batch. AAPS PharmSciTech 2020; 21:176. [PMID: 32572701 PMCID: PMC7308264 DOI: 10.1208/s12249-020-01713-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/13/2020] [Indexed: 12/11/2022] Open
Abstract
This paper presents a rational workflow for developing enabling formulations, such as amorphous solid dispersions, via hot-melt extrusion in less than a year. First, our approach to an integrated product and process development framework is described, including state-of-the-art theoretical concepts, modeling, and experimental characterization described in the literature and developed by us. Next, lab-scale extruder setups are designed (processing conditions and screw design) based on a rational, model-based framework that takes into account the thermal load required, the mixing capabilities, and the thermo-mechanical degradation. The predicted optimal process setup can be validated quickly in the pilot plant. Lastly, a transfer of the process to any GMP-certified manufacturing site can be performed in silico for any extruder based on our validated computational framework. In summary, the proposed workflow massively reduces the risk in product and process development and shortens the drug-to-market time for enabling formulations.
Collapse
Affiliation(s)
- Josip Matić
- Research Center Pharmaceutical Engineering GmbH, Graz, Austria
| | - Amrit Paudel
- Research Center Pharmaceutical Engineering GmbH, Graz, Austria.
- Institute for Process and Particle Engineering, Graz University of Technology, Graz, Austria.
| | - Hannes Bauer
- Research Center Pharmaceutical Engineering GmbH, Graz, Austria
| | | | | | - Johannes G Khinast
- Research Center Pharmaceutical Engineering GmbH, Graz, Austria.
- Institute for Process and Particle Engineering, Graz University of Technology, Graz, Austria.
| |
Collapse
|
48
|
Probing the Molecular-Level Interactions in an Active Pharmaceutical Ingredient (API) - Polymer Dispersion and the Resulting Impact on Drug Product Formulation. Pharm Res 2020; 37:94. [PMID: 32405662 DOI: 10.1007/s11095-020-02813-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 04/02/2020] [Indexed: 10/24/2022]
Abstract
PURPOSE An investigation of underlying mechanisms of API-polymer interaction patterns has the potential to provide valuable insights for selecting appropriate formulations with superior physical stability and processability. MATERIALS AND METHODS In this study, copovidone was used as a polymeric carrier for several model compounds including clotrimazole, nifedipine, and posaconazole. The varied chemical structures conferred the ability for the model compounds to form distinct interactions with copovidone. Rheology and nuclear magnetic resonance (NMR) were combined to investigate the molecular pattern and relative strength of active pharmaceutical ingredient (API)-polymer interactions. In addition, the impact of the interactions on formulation processability via hot melt extrusion (HME) and physical stability were evaluated. RESULTS The rheological response of an API-polymer system was found to be highly sensitive to API-polymer interaction, depending both on API chemistry and API-polymer miscibility. In the systems studied, dispersed API induced a stronger plasticizer effect on the polymer matrix compared to crystalline/aggregated API. Correspondingly, the processing torque via HME showed a proportional relationship with the maximum complex viscosity of the API-polymer system. In order to quantitatively evaluate the relative strength of the API-polymer interaction, homogeneously dispersed API-polymer amorphous samples were prepared by HME at an elevated temperature. DSC, XRD, and rheology were employed to confirm the amorphous integrity and homogeneity of the resultant extrudates. Subsequently, the homogeneously dispersed API-polymer amorphous dispersions were interrogated by rheology and NMR to provide a qualitative and quantitative assessment of the nature of the API-polymer interaction, both macroscopically and microscopically. Rheological master curves of frequency sweeps of the extrudates exhibited a strong dependence on the API chemistry and revealed a rank ordering of the relative strength of API-copovidone interactions, in the order of posaconazole > nifedipine > clotrimazole. NMR data provided the means to precisely map the API-polymer interaction pattern and identify the specific sites of interaction from a molecular perspective. Finally, the impact of API-polymer interactions on the physical stability of the resultant extrudates was studied. CONCLUSION Qualitative and quantitative evaluation of the relative strength of the API-polymer interaction was successfully accomplished by utilizing combined rheology and NMR. Graphical Abstract.
Collapse
|
49
|
Brinkmann J, Rest F, Luebbert C, Sadowski G. Solubility of Pharmaceutical Ingredients in Natural Edible Oils. Mol Pharm 2020; 17:2499-2507. [DOI: 10.1021/acs.molpharmaceut.0c00215] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Joscha Brinkmann
- Laboratory of Thermodynamics, TU Dortmund University, Emil-Figge-Str. 70, D-44227 Dortmund, Germany
| | - Frauke Rest
- Laboratory of Thermodynamics, TU Dortmund University, Emil-Figge-Str. 70, D-44227 Dortmund, Germany
| | - Christian Luebbert
- Laboratory of Thermodynamics, TU Dortmund University, Emil-Figge-Str. 70, D-44227 Dortmund, Germany
| | - Gabriele Sadowski
- Laboratory of Thermodynamics, TU Dortmund University, Emil-Figge-Str. 70, D-44227 Dortmund, Germany
| |
Collapse
|
50
|
|