1
|
Parkes A, Ziaee A, O'Reilly E. Evaluating experimental, knowledge-based and computational cocrystal screening methods to advance drug-drug cocrystal fixed-dose combination development. Eur J Pharm Sci 2024; 203:106931. [PMID: 39389169 DOI: 10.1016/j.ejps.2024.106931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/18/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
Fixed-dose combinations (FDCs) offer significant advantages to patients and the pharmaceutical industry alike through improved dissolution profiles, synergistic effects and extended patent lifetimes. Identifying whether two active pharmaceutical ingredients have the potential to form a drug-drug cocrystal (DDC) or interact is an essential step in determining the most suitable type of FDC to formulate. The lack of coherent strategies to determine if two active pharmaceutical ingredients that can be co-administered can form a cocrystal, has significantly impacted DDC commercialisation. This review aims to accelerate the development of FDCs and DDCs by evaluating existing experimental, knowledge-based and computational cocrystal screening methods; the background of their development, their application in screening for cocrystals and DDCs, and their limitations are discussed. The evaluation provided in this review will act as a guide for selecting suitable screening methods to accelerate FDC development.
Collapse
Affiliation(s)
- Alice Parkes
- Department of Chemical Sciences, SSPC the SFI Research Centre for Pharmaceuticals, Bernal Institute, University of Limerick, Limerick, Ireland
| | | | - Emmet O'Reilly
- Department of Chemical Sciences, SSPC the SFI Research Centre for Pharmaceuticals, Bernal Institute, University of Limerick, Limerick, Ireland.
| |
Collapse
|
2
|
Niederquell A, Herzig S, Schönenberger M, Stoyanov E, Kuentz M. Computational Support to Explore Ternary Solid Dispersions of Challenging Drugs Using Coformer and Hydroxypropyl Cellulose. Mol Pharm 2024; 21:5619-5631. [PMID: 39388157 PMCID: PMC11539070 DOI: 10.1021/acs.molpharmaceut.4c00592] [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: 05/29/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/15/2024]
Abstract
A majority of drugs marketed in amorphous formulations have a good glass-forming ability, while compounds less stable in the amorphous state still pose a formulation challenge. This work explores ternary solid dispersions of two model drugs with a polymer (i.e., hydroxypropyl cellulose) and a coformer as stabilizing excipients. The aim was to introduce a computational approach by preselecting additives using solubility parameter intervals (i.e., overlap range of solubility parameter, ORSP) followed by more advanced COSMO-RS theory modeling. Thus, a mapping of calculated mixing enthalpy and melting points is proposed for in silico evaluation prior to hot melt extrusion. Following experimental testing of process feasibility, the selected formulations were tested for their physical stability using conventional bulk analytics and by confocal laser scanning and atomic force microscopy imaging. In line with the in silico screening, dl-malic and l-tartaric acid (20%, w/w) in HPC formulations showed no signs of early drug crystallization after 3 months. However, l-tartaric acid formulations displayed few crystals on the surface, which was likely a humidity-induced surface phenomenon. Although more research is needed, the conclusion is that the proposed computational small-scale extrusion approach of ternary solid dispersion has great potential in the formulation development of challenging drugs.
Collapse
Affiliation(s)
- Andreas Niederquell
- Institute
for Pharma Technology, University of Applied
Sciences and Arts Northwestern Switzerland, School of Life Sciences
FHNW, Hofackerstr. 30, 4132 Muttenz, Switzerland
| | - Susanne Herzig
- Institute
for Pharma Technology, University of Applied
Sciences and Arts Northwestern Switzerland, School of Life Sciences
FHNW, Hofackerstr. 30, 4132 Muttenz, Switzerland
| | - Monica Schönenberger
- Nano
Imaging Lab, Swiss Nanoscience Institute, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Edmont Stoyanov
- Nisso
Chemical Europe, Berliner
Allee 42, 40212 Düsseldorf, Germany
| | - Martin Kuentz
- Institute
for Pharma Technology, University of Applied
Sciences and Arts Northwestern Switzerland, School of Life Sciences
FHNW, Hofackerstr. 30, 4132 Muttenz, Switzerland
| |
Collapse
|
3
|
Li Y, Wei Q, Su J, Zhang H, Fan Z, Ding Z, Wen M, Liu M, Zhao Y. Encapsulation of astaxanthin in OSA-starch based amorphous solid dispersions with HPMCAS-HF/Soluplus® as effective recrystallization inhibitor. Int J Biol Macromol 2024; 279:135421. [PMID: 39349321 DOI: 10.1016/j.ijbiomac.2024.135421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/05/2024] [Accepted: 09/05/2024] [Indexed: 10/02/2024]
Abstract
In this study, the interaction among multifunctional excipients, including polysaccharides, cellulose derivatives, and surfactants, was particularly investigated, together with its impact on the physicochemical properties of astaxanthin amorphous solid dispersions (ASTX ASDs). It was indicated that Span 20 could rapidly form hemimicelles or aggregates in the presence of hypromellose acetate succinate HF (HPMCAS-HF, HF) or Soluplus®, while octenyl succinic anhydride modified starch (OSA-starch) efficiently assisted in the coalescence inhibition of drug-excipients aggregates, which was jointly beneficial to the recrystallization inhibition of amorphous ASTX. ASTX ASDs were further prepared with OSA-starch, HPMCAS-HF/Soluplus®, and Span 20 as the wall materials. DSC, SEM, and XRD confirmed that crystalline ASTX had transformed to amorphous state in the ASDs, while FT-IR spectra provided evidence suggesting the existence of hydrogen bonds and hydrophobic interaction between ASTX and the excipients. The dissolution of ASTX ASDs in different media revealed significant promotion, while the pharmacokinetic results further demonstrated the oral bioavailability of ASTX ASDs enhanced remarkably, exhibiting 2.75-fold (SD1) and 1.87-fold (SD2) increase, respectively, compared to ASTX bulk powder. In summary, the cellulose derivatives-surfactant interaction had great impact on the physicochemical properties of ASTX ASDs, and their combinations exhibited great potential for delivering the hydrophobic bioactive compounds efficiently.
Collapse
Affiliation(s)
- Yinglan Li
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252059, People's Republic of China
| | - Qipeng Wei
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252059, People's Republic of China
| | - Jianshuo Su
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252059, People's Republic of China
| | - Huaizhen Zhang
- School of Geography and Environment, Liaocheng University, Liaocheng, Shandong 252059, People's Republic of China
| | - Zhiping Fan
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252059, People's Republic of China
| | - Zhuang Ding
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252059, People's Republic of China
| | - Min Wen
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252059, People's Republic of China
| | - Min Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252059, People's Republic of China
| | - Yanna Zhao
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252059, People's Republic of China.
| |
Collapse
|
4
|
Coutinho AL, Hom K, Polli JE. Prediction of Successful Amorphous Solid Dispersion Pairs through Liquid State Nuclear Magnetic Resonance. Mol Pharm 2024. [PMID: 39482969 DOI: 10.1021/acs.molpharmaceut.4c00359] [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: 11/03/2024]
Abstract
Amorphous solid dispersions (ASDs) function in part via a "parachute effect", i.e., polymer-enabled prolonged drug supersaturation, presumably through drug-polymer interactions in the liquid state. We aim to expand the utility of liquid state nuclear magnetic resonance (1HNMR) to streamline polymer selection for ASDs. Our hypothesis is that strong molecular interactions between polymer and drug in 1HNMR anticipate reduced precipitation kinetics in supersaturation studies. For three drug-polymer pairs (i.e., etravirine with each HPMC, HPMCAS-M, and PVP-VA), 1HNMR findings were compared to more common supersaturation studies. Drug-polymer interactions were assessed by saturation transfer difference NMR (STD-NMR) and T1 relaxation time. 2D-1H NOESY experiments were also performed. Supersaturation studies involved precipitation inhibition using the solvent-shift methodology. The results from STD-NMR and T1 relaxation time indicate etravirine bound preferably to HPMCAS-M > HPMC ≫ PVP-VA. STD-NMR and T1 relaxation time yielded insight into which fragments of etravirine structure bind with HPMCAS-M and HPMC. The strong interactions from STD-NMR and T1 relaxation time changes indicated that HPMCAS-M and HPMC, but not PVP-VA, are suitable polymers to maintain etravirine supersaturation and inhibit drug precipitation. 2D-1H NOESY results corroborate the findings of STD-NMR and T1 relaxation time, showing that etravirine interacts preferably to HPMCAS-M than to PVP-VA. Supersaturation studies using solvent-shift technique corroborated our hypothesis as predissolved HPMCAS-M and HPMC, but to a less extent PVP-VA, markedly promoted etravirine supersaturation and inhibited drug precipitation. Supersaturation studies agreed with STD-NMR and T1 relaxation time predictions, as HPMC and HPMCAS-M maintained etravirine in solution for longer time than PVP-VA. The results show promise of 1HNMR to streamline polymer selection in a nondestructive and resource sparing fashion for subsequent ASD development.
Collapse
Affiliation(s)
- Ana L Coutinho
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Kellie Hom
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - James E Polli
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| |
Collapse
|
5
|
Nespi M, Ly J, Fan Y, Chen S, Liu L, Gu Y, Castleberry S. Vehicle Effect on In-Vitro and In-Vivo Performance of Spray Dry Dispersions. J Pharm Sci 2024:S0022-3549(24)00491-X. [PMID: 39486520 DOI: 10.1016/j.xphs.2024.10.043] [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: 07/13/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024]
Abstract
In early drug development, amorphous spray-dried dispersions (SDDs) applied to enhance the bioavailability of poorly water-soluble compounds are typically administered to preclinical species via oral gavage in the form of suspensions. The liquid formulations are usually prepared on the same day of dosing to minimize the exposure of the amorphous material to the aqueous vehicle, thereby reducing the risk of crystallization. Dose-ability (e.g. syringeability) of the suspensions is also a critical factor for the administration, particularly when high doses, thus concentrations, are required for toxicology studies. As a result, it is standard practice during early formulation screening to assess the stability and the maximum feasible concentration of SDDs in various vehicles. In this study, we evaluated the impact of different vehicles on the performance of a model SDD in in-vitro and in-vivo settings, to mitigate the risks associated with its administration in liquid form. A poorly water-soluble compound (GEN-A) was selected to screen various SDDs and generate the SDD model at 30% drug load with HPMCAS-MF polymer carrier. The SDD was suspended in selected aqueous vehicles after a careful vehicle components screening, that included suspending agents (HPC-SL), solubilizers (PEG400, Propylene glycol), surfactants (Vitamin E TPGS, SLS, Tween 80, Poloxamer 188), and complexing agents (HP-ꞵ-CD, SBE-ꞵ-CD). The suspensions were characterized for stability, dose-ability and dissolution in biorelevant media, prior administration in pre-clinical species. The SDD dissolution profile revealed that the drug's supersaturation level was positively impacted by the presence of a surfactant (SLS) and a complexing agent (SBE-ꞵ-CD) with respect to a suspending agents (HPC-SL) in the vehicle. Similarly, the pharmacokinetics profiles of the drug following the administration of the SDD in a vehicle with a complexing agent (SBE-ꞵ-CD) achieved greater exposure compare to the SDD in a vehicle with a suspending agent (HPC-SL). These findings confirm a synergistic effect between the SDD and the vehicles, suggesting that this combination could be leveraged to maximize the advantages of the amorphous approach.
Collapse
Affiliation(s)
- Marika Nespi
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA.
| | - Justin Ly
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Yuchen Fan
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Shu Chen
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Liling Liu
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Yimin Gu
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| | - Steven Castleberry
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California, 94080, USA
| |
Collapse
|
6
|
Novick A, Cai D, Nguyen Q, Garnett R, Adams R, Toberer E. Probabilistic prediction of material stability: integrating convex hulls into active learning. MATERIALS HORIZONS 2024; 11:5381-5393. [PMID: 39158003 DOI: 10.1039/d4mh00432a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Active learning is a valuable tool for efficiently exploring complex spaces, finding a variety of uses in materials science. However, the determination of convex hulls for phase diagrams does not neatly fit into traditional active learning approaches due to their global nature. Specifically, the thermodynamic stability of a material is not simply a function of its own energy, but rather requires energetic information from all other competing compositions and phases. Here we present convex hull-aware active learning (CAL), a novel Bayesian algorithm that chooses experiments to minimize the uncertainty in the convex hull. CAL prioritizes compositions that are close to or on the hull, leaving significant uncertainty in other compositions that are quickly determined to be irrelevant to the convex hull. The convex hull can thus be predicted with significantly fewer observations than approaches that focus solely on energy. Intrinsic to this Bayesian approach is uncertainty quantification in both the convex hull and all subsequent predictions (e.g., stability and chemical potential). By providing increased search efficiency and uncertainty quantification, CAL can be readily incorporated into the emerging paradigm of uncertainty-based workflows for thermodynamic prediction.
Collapse
Affiliation(s)
- Andrew Novick
- Department of Physics, Colorado School of Mines, Golden, Colorado, USA.
| | - Diana Cai
- Center for Computational Mathematics, Flatiron Institute Address, New York, New York, USA
| | - Quan Nguyen
- Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Roman Garnett
- Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Ryan Adams
- Department of Computer Science, Princeton University, New Jersey, USA
| | - Eric Toberer
- Department of Physics, Colorado School of Mines, Golden, Colorado, USA.
| |
Collapse
|
7
|
Mathers A, Fulem M. Drug-polymer compatibility prediction via COSMO-RS. Int J Pharm 2024; 664:124613. [PMID: 39179010 DOI: 10.1016/j.ijpharm.2024.124613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/16/2024] [Accepted: 08/17/2024] [Indexed: 08/26/2024]
Abstract
In this work, the solid-liquid equilibrium (SLE) curve for ten active pharmaceutical ingredients (APIs) with the polymer polyvinylpyrrolidone (PVP) K12 was purely predicted using the Conductor-like Screening Model for Real Solvents (COSMO-RS). In particular, two COSMO-RS-based strategies were followed (i.e., a traditional approach and an expedited approach), and their performances were compared. The veracity of the predicted SLE curves was assessed via a comparison with their respective SLE dataset that was obtained using the step-wise dissolution (S-WD) method. Overall, the COSMO-RS-based API-PVP K12 SLE curves were in satisfactory agreement with the S-WD-based data points. Of the twenty predicted SLE curves, only two were found to be in strong disagreement with the corresponding experimental values (both modeled using the expedited approach). Hence, it was recommended to use the traditional approach when predicting the API-polymer SLE curve. At the present moment, COSMO-RS may be an effective computational tool for the expeditious screening of API-polymer compatibility, particularly in the case of promising novel APIs, for which experimental datasets are likely limited or non-existent.
Collapse
Affiliation(s)
- Alex Mathers
- 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
|
8
|
Indulkar AS, Alex S, Zhang GGZ. Impact of dissolution medium pH and ionization state of the drug on the release performance of amorphous solid dispersions. J Pharm Sci 2024:S0022-3549(24)00478-7. [PMID: 39454946 DOI: 10.1016/j.xphs.2024.10.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Amorphous solid dispersions (ASDs) are widely employed as a strategy to improve oral bioavailability of poorly water soluble compounds. Typically, optimal dissolution performance from a polyvinylpyrrolidone vinyl acetate (PVPVA) based ASD is observed at relatively low drug loading limit. Above a certain drug load, termed limit of congruency (LoC), the release from ASDs significantly decreases. So far, the majority of the dissolution behavior has been tested in conditions where the drug primarily exists in unionized form. In this work, the impact of pH of the dissolution environment on the release performance of ASDs of an ionizable drug was studied. Atazanavir (ATZ), a weakly basic drug with a pKa of 4.5 was used as a model compound and PVPVA was used as a non-ionizable matrix polymer. Dissolution rate was measured using Wood's apparatus which normalizes the surface area of the dissolving tablet. The pH of the dissolution media was varied between 1 and 6.8, to cover a range where ATZ exists as >99 % ionized or unionized species. At pH 6.8, near complete release was observed only when the drug load was ≤ 6 %. Unlike typically observed drastic decline in release behavior for PVPVA based ASDs above LoC, ATZ ASDs underwent gradual decline in dissolution behavior when the DL was increased to 8 %. This was attributed to potential formation of an ATZ-PVPVA associated phase with dissolution rate slower than neat PVPVA. However, the 10 % DL ASD showed negligible ATZ release. On another extreme (pH 1) where ATZ is ∼100 % ionized, the dissolution rate of ATZ was faster than that of PVPVA. ASD dissolution rate was found to be slower than that of the neat drug but faster than PVPVA and interestingly, did not change with DL. This can be attributed to formation of an ionized ATZ-PVPVA phase which controls the dissolution rate of the ASD. At pH 3, where the drug is ∼97 % ionized, near complete release was observed for drug loads ≤ 8 %. To observe significant increase in drug loading with near complete release, >98 % ionization of ATZ was required. At pH 2 where ATZ is ∼99.7 % ionized, near complete release was observed for drug loads up to 30 %. Furthermore, the deterioration in dissolution performance with an increase in drug load continued to be gradual at pH 2. The enhancement in dissolution performance did not correlate with solubility enhancement of ATZ due to ionization. We theorize that the enhancement in the dissolution performance due to ionization is the result of formation of an ionized ATZ-PVPVA phase which increases the hydrophilicity and the miscibility of the ASD. This can help resist water induced phase separation during ASD dissolution and therefore, result in continuous, and congruent dissolution of the drug and polymer.
Collapse
Affiliation(s)
- Anura S Indulkar
- Small Molecule CMC Development, Research and Development, AbbVie Inc., North Chicago, IL 60064, United States.
| | - Samantha Alex
- Small Molecule CMC Development, Research and Development, AbbVie Inc., North Chicago, IL 60064, United States
| | - Geoff G Z Zhang
- Small Molecule CMC Development, Research and Development, AbbVie Inc., North Chicago, IL 60064, United States
| |
Collapse
|
9
|
Sherif AY, Abbas Ibrahim M. Self-Nanoemulsifying Drug Delivery System Combined with a Polymeric Amorphous System of Glibenclamide for Enhanced Drug Dissolution and Stability. ACS OMEGA 2024; 9:43165-43174. [PMID: 39464452 PMCID: PMC11500158 DOI: 10.1021/acsomega.4c07285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/28/2024] [Accepted: 10/04/2024] [Indexed: 10/29/2024]
Abstract
Self-nanoemulsifying drug delivery systems (SNEDDS) have been widely applied to improve the dissolution and bioavailability of hydrophobic medications like glibenclamide (GB). However, the acid liability of GB limits its loading in SNEDDS formulation owing to the expected drug degradation. The present study investigated the ability of a polymeric amorphous system (PAS) to amorphize raw GB and facilitate its integration within dispersed SNEDDS. Liquid-SNEDDS (L-SNEDDS), solid-SNEDDS (S-SNEDDS), and combined systems (SNEDDS + PAS) were prepared for this purpose. The physicochemical properties of the prepared formulations were examined using a zeta-sizer, SEM, DSC, PXRD, and dissolution apparatus. In addition, GB integrity within formulations following incubation in a stability chamber was also investigated. The prepared formulations were able to be dispersed within the nanosize range. SEM, DSC, and PXRD showed that freeze-drying (FD) was superior to the microwave (MW) method in GB amorphization. Even though L-SNEDDS and S-SNEDDS were able to increase the dissolution efficiency (DE) of GB, drug degradation was observed. However, PAS prepared using FD was able to increase the DE of GB from 2.5% to 84.2% and protect the drug from chemical degradation. The present study revealed that a combined system (SNEDDS + PAS) is a promising approach to enhance the stability of acid-labile drugs and facilitate the integration of amorphous drugs within a dispersed SNEDDS formulation.
Collapse
Affiliation(s)
- Abdelrahman Y. Sherif
- Department of Pharmaceutics,
College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohamed Abbas Ibrahim
- Department of Pharmaceutics,
College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
10
|
Kossor C, Bhat R, Davé RN. Assessing processability of milled HME extrudates: Consolidating the effect of extrusion temperature, drug loading, and particle size via Non-dimensional cohesion. Int J Pharm 2024; 666:124833. [PMID: 39414188 DOI: 10.1016/j.ijpharm.2024.124833] [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: 08/08/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024]
Abstract
The downstream processability of Hot Melt Extrusion (HME) Amorphous Solid Dispersions (ASD), an underexplored topic of importance, was assessed through a multi-faceted particle engineering approach. Extrudates, comprised of griseofulvin (GF), a model poorly water-soluble drug, and hydroxypropyl cellulose (HPC), were prepared at four drug concentrations and three HME temperature profiles to yield cases with and without residual crystallinity and subsequently milled to five sieve cuts ranging from < 45 μm to 355 - 500 μm. Solid state characterization was performed with XRPD, FT-IR, and TGA. Particle scale properties of the milled extrudates were evaluated including particle size, density, surface energy, and morphologies imaged via SEM. It was observed that regardless of sieve cut size, drug concentration and HME conditions impacted the flowability trends, quantified via Flow Function Coefficient (FFC) and bulk density. As a novelty, the effects of various process parameters and drug loadings were consolidated into a dimensionless interparticle cohesion measure, granular Bond Number (Bog), to better correlate them with bulk powder properties. The significant contrast in particle morphologies, particle size, and densities among selected cases demonstrated that particle size alone should not be the sole consideration when correlating particle scale to bulk powder scale properties of milled extrudates. Instead, the HME temperature profile and ASD drug loading may be more suitable parameters affecting the bulk powder properties of the milled extrudates.
Collapse
Affiliation(s)
- Christopher Kossor
- New Jersey Center for Engineered Particulates, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Roopal Bhat
- New Jersey Center for Engineered Particulates, New Jersey Institute of Technology, Newark, NJ 07102, USA
| | - Rajesh N Davé
- New Jersey Center for Engineered Particulates, New Jersey Institute of Technology, Newark, NJ 07102, USA.
| |
Collapse
|
11
|
Polyzois H, Nguyen HT, Roberto de Alvarenga Junior B, Taylor LS. Amorphous Solid Dispersion Formation for Enhanced Release Performance of Racemic and Enantiopure Praziquantel. Mol Pharm 2024; 21:5285-5296. [PMID: 39292641 PMCID: PMC11462518 DOI: 10.1021/acs.molpharmaceut.4c00711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/20/2024]
Abstract
Praziquantel (PZQ) is the treatment of choice for schistosomiasis, which affects more than 250 million people globally. Commercial tablets contain the crystalline racemic compound (RS-PZQ) which limits drug dissolution and oral bioavailability and can lead to unwanted side effects and poor patient compliance due to the presence of the S-enantiomer. While many approaches have been explored for improving PZQ's dissolution and oral bioavailability, studies focusing on investigating its release from amorphous solid dispersions (ASDs) have been limited. In this work, nucleation induction time experiments were performed to identify suitable polymers for preparing ASDs using RS-PZQ and R-PZQ, the therapeutically active enantiomer. Cellulose-based polymers, hydroxypropyl methylcellulose acetate succinate (HPMCAS, MF grade) and hydroxypropyl methylcellulose (HPMC, E5 LV grade), were the best crystallization inhibitors for RS-PZQ in aqueous media and were selected for ASD preparation using solvent evaporation (SE) and hot-melt extrusion (HME). ASDs prepared experimentally were subjected to X-ray powder diffraction to verify their amorphous nature and a selected number of ASDs were monitored and found to remain physically stable following several months of storage under accelerated-stability testing conditions. SE HPMCAS-MF ASDs of RS-PZQ and R-PZQ showed faster release than HPMC E5 LV ASDs and maintained good performance with an increase in drug loading (DL). HME ASDs of RS-PZQ formulated using HPMCAS-MF exhibited slightly enhanced release compared to that of SE ASDs. SE HPMCAS-MF ASDs showed a maximum release increase of the order of 6 times compared to generic and branded (Biltricide) PZQ tablets. More importantly, SE R-PZQ ASDs with HPMCAS-MF released the drug as effectively as RS-PZQ or better, depending on the DL used. These findings have significant implications for the development of commercial PZQ formulations comprised solely of the R-enantiomer, which can result in mitigation of the biopharmaceutical and compliance issues associated with current commercial tablets.
Collapse
Affiliation(s)
- Hector Polyzois
- Department of Industrial and Molecular
Pharmaceutics, College of Pharmacy, Purdue
University, West Lafayette, Indiana 47907, United States
| | - Hanh Thuy Nguyen
- Department of Industrial and Molecular
Pharmaceutics, College of Pharmacy, Purdue
University, West Lafayette, Indiana 47907, United States
| | | | - Lynne S. Taylor
- Department of Industrial and Molecular
Pharmaceutics, College of Pharmacy, Purdue
University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
12
|
Zhang W, Thool P, Weitz BW, Hou HH. Investigating the effects of formulation variables on the disintegration of spray dried amorphous solid dispersion tablets. J Pharm Sci 2024:S0022-3549(24)00432-5. [PMID: 39374694 DOI: 10.1016/j.xphs.2024.09.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/09/2024]
Abstract
Amorphous solid dispersion (ASD) tablets based on hydrophilic polymer carriers may encounter disintegration challenges. In this work, the effect of different formulation composition variables on the ASD tablet disintegration performance was systematically studied. GDC-0334: copovidone (PVPVA) 60: 40 ASD prepared by spray drying was selected as the model ASD system. The effects of ASD loading, filler type and ratio, disintegrant type and level were then investigated using tablets made by direct compression process. Tablet disintegration time increased with the increase of ASD loading, especially when ASD loading exceeded 50 %. At the same tablet solid fraction, when lactose was used as the soluble filler, faster tablet disintegration was observed compared to the tablets with mannitol as the soluble filler. Among the three tested disintegrants, croscarmellose sodium performed the best in facilitating the ASD tablet disintegration, followed by sodium starch glycolate, and crospovidone was the poorest. When croscarmellose sodium was used as the disintegrant, 5 % level was sufficient to enable ASD tablet disintegration at 60 % ASD loading and further increase of croscarmellose sodium level to 8 % did not provide additional benefit. Water uptake experiments were performed on selected tablets and the results demonstrated a positive correlation with tablet disintegration time, indicating water penetration is a major contributing step for the disintegration of our ASD tablets. Overall, this work provides a rationale for excipient selection and insights into building a platform formulation approach for developing immediate-release ASD tablets.
Collapse
Affiliation(s)
- Wei Zhang
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Prajwal Thool
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Benjamin W Weitz
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hao Helen Hou
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
13
|
Liu L, Wang Y, Sun J, Zhang Y, Zhang X, Wu L, Liu Y, Zhang X, Xia Y, Zhang Q, Gao N. Improved photostability, solubility, hygroscopic stability and antimicrobial activity of fleroxacin by synthesis of fleroxacin-D-tartaric acid pharmaceutical salt. Eur J Pharm Biopharm 2024; 203:114464. [PMID: 39181416 DOI: 10.1016/j.ejpb.2024.114464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 08/09/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
To improve the solubility of the fluoroquinolone drug fleroxacin (FL), based on the previous experience of our research group in synthesizing co-crystals/salts of quinolone drugs to improve the physicochemical properties of drugs, Fleroxacin-D-tartaric acid dihydrate salt (FL-D-TT, C17H19F3N3O3·C4H5O6·2(H2O)), was synthesized for the first time using fleroxacin and D/L-tartaric acid (D/L-TT). Structural characterization of FL-D-TT was carried out using single-crystal X-ray diffraction, infrared spectral analysis (FT-IR) and powder X-ray diffraction (PXRD). Molecular electrostatic potential analysis showed that D-tartaric acid interacted more readily with FL than L-tartaric acid. The solubility of FL-D-TT (9.71 mg/mL, 1.82 mg/mL) was significantly higher compared to FL (0.39 mg/mL, 0.71 mg/mL) in water and buffer solution at pH 7.4. This may be attributed to the formation of charge-assisted hydrogen bonds (CAHBs) between FL and D-TT that facilitates the dissociation of FL cations in the dissolution medium, leading to an increase in FL solubility. This also led to some improvement in the in vitro antimicrobial activity of FL-D-TT against E. coli, S. typhi, and S. aureus. In addition, the hygroscopic stability of FL has been improved. Surprisingly, FL-D-TT had better photostability than FL, which could be attributed to the introduction of D-TT to make the photosensitizing moiety of FL more stable, which led to the improvement of the photostability of FL.
Collapse
Affiliation(s)
- Lixin Liu
- College of Pharmacy, Jiamusi University, Heilongjiang Province, Jiamusi 154007, China.
| | - Yuning Wang
- College of Pharmacy, Jiamusi University, Heilongjiang Province, Jiamusi 154007, China
| | - Jiuyi Sun
- College of Pharmacy, Jiamusi University, Heilongjiang Province, Jiamusi 154007, China
| | - Yunan Zhang
- College of Pharmacy, Jiamusi University, Heilongjiang Province, Jiamusi 154007, China.
| | - Xiangyu Zhang
- College of Pharmacy, Jiamusi University, Heilongjiang Province, Jiamusi 154007, China
| | - Lili Wu
- College of Pharmacy, Jiamusi University, Heilongjiang Province, Jiamusi 154007, China
| | - Yingli Liu
- College of Pharmacy, Jiamusi University, Heilongjiang Province, Jiamusi 154007, China
| | - Xuan Zhang
- College of Pharmacy, Jiamusi University, Heilongjiang Province, Jiamusi 154007, China
| | - Yidi Xia
- College of Pharmacy, Jiamusi University, Heilongjiang Province, Jiamusi 154007, China
| | - Qiumei Zhang
- College of Pharmacy, Jiamusi University, Heilongjiang Province, Jiamusi 154007, China
| | - Ning Gao
- College of Pharmacy, Jiamusi University, Heilongjiang Province, Jiamusi 154007, China
| |
Collapse
|
14
|
Alvebratt C, Karlén F, Åhlén M, Edueng K, Dubbelboer I, Bergström CAS. Benefits of combining supersaturating and solubilizing formulations - Is two better than one? Int J Pharm 2024; 663:124437. [PMID: 39002818 DOI: 10.1016/j.ijpharm.2024.124437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/15/2024]
Abstract
A variety of enabling formulations has been developed to address poor oral drug absorption caused by insufficient dissolution in the gastrointestinal tract. As the in vivo performance of these formulations is a result of a complex interplay between dissolution, digestion and permeation, development of suitable in vitro assays that captures these phenomena are called for. The enabling-absorption (ENA) device, consisting of a donor and receiver chamber separated by a semipermeable membrane, has successfully been used to study the performance of lipid-based formulations. In this work, the ENA device was prepared with two different setups (a Caco-2 cell monolayer and an artificial lipid membrane) to study the performance of a lipid-based formulation (LBF), an amorphous solid dispersion (ASD) and the potential benefit of combining the two formulation strategies. An in vivo pharmacokinetic study in rats was performed to evaluate the in vitro-in vivo correlation. In the ENA, high drug concentrations in the donor chamber did not translate to a high mass transfer, which was particularly evident for the ASD as compared to the LBF. The solubility of the polymer used in the ASD was strongly affected by pH-shifts in vitro, and the ph_dependence resulted in poor in vivo performance of the formulation. The dissolution was however increased in vitro when the ASD was combined with a blank lipid-based formulation. This beneficial effect was also observed in vivo, where the drug exposure of the ASD increased significantly when the ASD was co-administered with the blank LBF. To conclude, the in vitro model managed to capture solubility limitations and strategies to overcome these for one of the formulations studied. The correlation between the in vivo exposure of the drug exposure and AUC in the ENA was good for the non pH-sensitive formulations. The deconvoluted pharmacokinetic data indicated that the receiver chamber was a better predictor for the in vivo performance of the drug, however both chambers provided valuable insights to the observed outcome in vivo. This shows that the advanced in vitro setting used herein successfully could explain absorption differences of highly complex formulations.
Collapse
Affiliation(s)
- Caroline Alvebratt
- Department of Pharmacy, Uppsala Biomedical Centre, P.O. Box 580, Uppsala University, Uppsala SE-751 23, Sweden.
| | - Filip Karlén
- Department of Pharmacy, Uppsala Biomedical Centre, P.O. Box 580, Uppsala University, Uppsala SE-751 23, Sweden.
| | - Michelle Åhlén
- Division of Nanotechnology and Functional Materials, Department of Engineering Sciences, Uppsala University, Uppsala SE-75121, Sweden.
| | - Khadijah Edueng
- Department of Pharmacy, Uppsala Biomedical Centre, P.O. Box 580, Uppsala University, Uppsala SE-751 23, Sweden
| | - Ilse Dubbelboer
- The Swedish Drug Delivery Center, Department of Pharmaceutical Biosciences, Uppsala Biomedical Centre, P.O. Box 591, Uppsala University, Uppsala SE-751 24, Sweden.
| | - Christel A S Bergström
- Department of Pharmacy, Uppsala Biomedical Centre, P.O. Box 580, Uppsala University, Uppsala SE-751 23, Sweden; The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala Biomedical Centre, P.O. Box 580, Uppsala University, Uppsala SE-751 23, Sweden.
| |
Collapse
|
15
|
Dos Santos KA, Chaves LL, Nadvorny D, de La Roca Soares MF, Sobrinho JLS. Exploring Co-Amorphous Formulations Of Nevirapine: Insights From Computational, Thermal, And Solubility Analyses. AAPS PharmSciTech 2024; 25:214. [PMID: 39266781 DOI: 10.1208/s12249-024-02932-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/03/2024] [Indexed: 09/14/2024] Open
Abstract
This study aimed to assess the formation of nevirapine (NVP) co-amorphs systems (CAM) with different co-formers (lamivudine-3TC, citric acid-CAc, and urea) through combined screening techniques as computational and thermal studies, solubility studies; in addition to develop and characterize suitable NVP-CAM. NVP-CAM were obtained using the quench-cooling method, and characterized by differential scanning calorimetry (DSC), X-ray diffractometry (XRD), Fourier Transform Infrared Spectroscopy (FTIR), and polarized light microscopy (PLM), in addition to in vitro dissolution in pH 6.8. The screening results indicated intermolecular interactions occurring between NVP and 3TC; NVP and CAc, where shifts in the melting temperature of NVP were verified. The presence of CAc impacted the NVP equilibrium solubility, due to hydrogen bonds. DSC thermograms evidenced the reduction and shifting of the endothermic peaks of NVP in the presence of its co-formers, suggesting partial miscibility of the compounds. Amorphization was proven by XRD and PLM assays. In vitro dissolution study exhibited a significant increase in solubility and dissolution efficiency of NVP-CAM compared to free NVP. Combined use of screening studies was useful for the development of stable and amorphous NVP-CAM, with increased NVP solubility, making CAM promising systems for combined antiretroviral therapy.
Collapse
Affiliation(s)
- Kayque Almeida Dos Santos
- Quality Control Core of Medicines and Correlates - NCQMC, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, PE, Brazil
| | - Luíse Lopes Chaves
- Quality Control Core of Medicines and Correlates - NCQMC, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, PE, Brazil
- Postgraduate Program in Pharmaceutical Sciences, Pharmaceutical Sciences Institute, Federal University of Alagoas, Maceió, Alagoas, Brazil
| | - Daniela Nadvorny
- Postgraduate Program in Pharmaceutical Sciences, Pharmaceutical Sciences Institute, Federal University of Alagoas, Maceió, Alagoas, Brazil
| | - Mônica Felts de La Roca Soares
- Quality Control Core of Medicines and Correlates - NCQMC, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, PE, Brazil
- Postgraduate Program in Pharmaceutical Sciences, Pharmaceutical Sciences Institute, Federal University of Alagoas, Maceió, Alagoas, Brazil
| | - José Lamartine Soares Sobrinho
- Quality Control Core of Medicines and Correlates - NCQMC, Department of Pharmaceutical Sciences, Federal University of Pernambuco, Recife, PE, Brazil.
| |
Collapse
|
16
|
Mohamed EM, Dharani S, Khuroo T, Nutan MTH, Cook P, Arunagiri R, Khan MA, Rahman Z. Oral Bioavailability Enhancement of Poorly Soluble Drug by Amorphous Solid Dispersion Using Sucrose Acetate Isobutyrate. AAPS PharmSciTech 2024; 25:202. [PMID: 39237685 DOI: 10.1208/s12249-024-02924-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/19/2024] [Indexed: 09/07/2024] Open
Abstract
The focus of the present work was to develop amorphous solid dispersion (ASD) formulation of aprepitant (APT) using sucrose acetate isobutyrate (SAIB) excipient, evaluate for physicochemical attributes, stability, and bioavailability, and compared with hydroxypropyl methylcellulose (HPMC) based formulation. Various formulations of APT were prepared by solvent evaporation method and characterized for physiochemical and in-vivo performance attributes such as dissolution, drug phase, stability, and bioavailability. X-ray powder diffraction indicated crystalline drug conversion into amorphous phase. Dissolution varied as a function of drug:SAIB:excipient proportion. The dissolution was more than 80% in the optimized formulation (F10) and comparable to HPMC based formulation (F13). Stability of F10 and F13 formulations stored at 25 C/60% and 40°C/75% RH for three months were comparable. Both ASD formulations (F10 and F13) were bioequivalent as indicated by the pharmacokinetic parameters Cmax and AUC0-∞. Cmax and AUC0-∞ of F10 and F13 formulations were 2.52 ± 0.39, and 2.74 ± 0.32 μg/ml, and 26.59 ± 0.39, and 24.79 ± 6.02 μg/ml.h, respectively. Furthermore, the bioavailability of ASD formulation was more than twofold of the formulation containing crystalline phase of the drug. In conclusion, stability and oral bioavailability of SAIB based ASD formulation is comparable to HPMC-based formulation of poorly soluble drugs.
Collapse
Affiliation(s)
- Eman M Mohamed
- Irma Lerma Rangel School of Pharmacy, Texas A&M Health Science Center, Texas A&M University, 310 Reynolds Medical Sciences Building, College Station, Texas, 77843-1114, U.S.A
- Department of Pharmaceutics, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Sathish Dharani
- Irma Lerma Rangel School of Pharmacy, Texas A&M Health Science Center, Texas A&M University, 310 Reynolds Medical Sciences Building, College Station, Texas, 77843-1114, U.S.A
| | - Tahir Khuroo
- Irma Lerma Rangel School of Pharmacy, Texas A&M Health Science Center, Texas A&M University, 310 Reynolds Medical Sciences Building, College Station, Texas, 77843-1114, U.S.A
| | - Mohammad T H Nutan
- Irma Lerma Rangel School of Pharmacy, Texas A&M Health Science Center, Texas A&M University, Kingsville, Texas, 78363, U.S.A
| | - Phillip Cook
- Eastman Chemical Company, Kingsport, Tennessee, 37662, U.S.A
| | | | - Mansoor A Khan
- Irma Lerma Rangel School of Pharmacy, Texas A&M Health Science Center, Texas A&M University, 310 Reynolds Medical Sciences Building, College Station, Texas, 77843-1114, U.S.A
| | - Ziyaur Rahman
- Irma Lerma Rangel School of Pharmacy, Texas A&M Health Science Center, Texas A&M University, 310 Reynolds Medical Sciences Building, College Station, Texas, 77843-1114, U.S.A..
| |
Collapse
|
17
|
Lohmann CA, Bochmann E, Kyeremateng SO. Impact of surfactant raw material variability on extrudate clarity appearance (transparency) in HME continuous manufacturing. Pharm Dev Technol 2024; 29:684-690. [PMID: 38995216 DOI: 10.1080/10837450.2024.2378333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 06/09/2024] [Accepted: 07/06/2024] [Indexed: 07/13/2024]
Abstract
The appearance of an extrudate formulation was monitored during hot-melt extrusion (HME) continuous manufacturing over 3 days. The formulation matrix consisted of a polymeric component, copovidone, and a low molecular weight surfactant, polysorbate 80. Based on studies prior to the continuous manufacturing, the desired appearance of the target extrudate is translucent. Although process parameters such as feed rate and screw speed were fixed during the continuous manufacturing, the extrudate appearance changed over time from turbid to translucent. For root-cause investigation, the extrudates were analyzed offline by differential scanning calorimetry (DSC) and advanced polymer chromatography (APC™). Although the polysorbate 80 content of both turbid and translucent extrudates was within target, the glass transition temperature of the turbid extrudate was 2 °C above expected value. The observed turbidity was traced to lot-to-lot variability of the polysorbate 80 used in the continuous manufacturing, where APC™ analysis revealed that the relative content of the low molecular weight component varied from 23% to 27% in correlation with the evolution from turbid to translucent extrudates. This work stresses the importance of taking feeding material variability into account during continuous manufacturing.
Collapse
|
18
|
Chen W, Yan A, Sun T, Wang X, Sun W, Pan B. Self-nanomicellizing solid dispersion: A promising platform for oral drug delivery. Colloids Surf B Biointerfaces 2024; 241:114057. [PMID: 38924852 DOI: 10.1016/j.colsurfb.2024.114057] [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: 04/12/2024] [Revised: 06/18/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024]
Abstract
Amorphous solid dispersion (ASD) has been widely used to enhance the oral bioavailability of water-insoluble drugs for oral delivery because of its advantages of enhancing solubility and dissolution rate. However, the problems related to drug recrystallization after drug dissolution in media or body fluid have constrained its application. Recently, a self-nanomicellizing solid dispersion (SNMSD) has been developed by incorporating self-micellizing polymers as carriers to settle the problems, markedly improving the ability of supersaturation maintenance and enhancing the oral bioavailability of drug. Spontaneous formation and stability of the self-nanomicelle (SNM) have been proved to be the key to supersaturation maintenance of SNMSD system. This offers a novel research direction for maintaining supersaturation and enhancing the bioavailability of ASDs. To delve into the advantages of SNMSDs, we provide a concise review introducing the formation mechanism, characterization methods and stability of SNMs, emphasizing the advantages of SNMSDs for oral drug delivery facilitated by SNM formation, and discussing relevant research prospects.
Collapse
Affiliation(s)
- Weitao Chen
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuan Ming Yuan West Road, Hai Dian District, Beijing 100193, China
| | - An Yan
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuan Ming Yuan West Road, Hai Dian District, Beijing 100193, China
| | - Tiancong Sun
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuan Ming Yuan West Road, Hai Dian District, Beijing 100193, China
| | - Xu Wang
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuan Ming Yuan West Road, Hai Dian District, Beijing 100193, China
| | - Weiwei Sun
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuan Ming Yuan West Road, Hai Dian District, Beijing 100193, China.
| | - Baoliang Pan
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuan Ming Yuan West Road, Hai Dian District, Beijing 100193, China.
| |
Collapse
|
19
|
Baumgartner A, Planinšek O. Development of Orodispersible Tablets with Solid Dispersions of Fenofibrate and Co-Processed Mesoporous Silica for Improved Dissolution. Pharmaceutics 2024; 16:1060. [PMID: 39204405 PMCID: PMC11359594 DOI: 10.3390/pharmaceutics16081060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/05/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024] Open
Abstract
Poor water solubility is an important challenge in the development of oral patient-friendly solid dosage forms. This study aimed to prepare orodispersible tablets with solid dispersions of a poorly water-soluble drug fenofibrate and a co-processed excipient consisting of mesoporous silica and isomalt. This co-processed excipient, developed in a previous study, exhibited improved flow and compression properties compared to pure silica while maintaining a high specific surface area for drug adsorption. Rotary evaporation was used to formulate solid dispersions with different amounts of fenofibrate, which were evaluated for solid state properties and drug release. The solid dispersion with 30% fenofibrate showed no signs of crystallinity and had a significantly improved dissolution rate, making it the optimal sample for formulation or orodispersible tablets. The aim was to produce tablets with minimal amounts of additional excipients while achieving a drug release profile similar to the uncompressed solid dispersion. The compressed formulations met the requirements for orodispersible tablets in terms of disintegration time, and the drug release from best formulation approximated the profile of uncompressed solid dispersion. Future research should focus on reducing the disintegration time and tablet size to enhance patient acceptability further.
Collapse
Affiliation(s)
- Ana Baumgartner
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Odon Planinšek
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| |
Collapse
|
20
|
Seo S, Kim GY, Kim MH, Lee KW, Kim MJ, Chaudhary M, Bikram K, Kim T, Choi S, Yang H, Park JW, Kim DD, Kim KT. Nanocrystal Formulation to Enhance Oral Absorption of Silybin: Preparation, In Vitro Evaluations, and Pharmacokinetic Evaluations in Rats and Healthy Human Subjects. Pharmaceutics 2024; 16:1033. [PMID: 39204378 PMCID: PMC11359960 DOI: 10.3390/pharmaceutics16081033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Despite the various therapeutic benefits and high tolerance of orally administered silybin, poor water-solubility can be the main restrictive physicochemical feature, which results in low oral bioavailability in the absorption. A milk thistle nanocrystal formulation (HM40) was prepared using a modified wet-milling method. Comprehensive characterization was performed to determine the physical morphology, crystallinity, and physicochemical properties. The long-term stability was evaluated over 24 months. In vitro silybin release was assessed at pH 1.2 for 2 h, followed by pH 6.8 for 4 h. Finally, in vivo pharmacokinetic studies were conducted in rats and healthy human volunteers. HM40 exhibited a nanocrystal structure maintaining crystallinity and enhanced the solubility and dissolution of silybin compared to that of the raw material. The stability over 24 months revealed consistent surface morphology, particle size, silybin content, and solubility. In vitro release profiles indicated a significant increase in the silybin release from HM40. In vivo pharmacokinetic studies demonstrated that HM40 showed 2.61- and 1.51-fold higher oral bioavailability in rats and humans, respectively, than that of the reference capsule. HM40 formulation presents a stable and promising approach for the oral delivery of poorly water-soluble silybin, with the potential for use in pharmaceutical formulations containing milk thistle.
Collapse
Affiliation(s)
- SeungRee Seo
- Life Science Research Institute, Daewoong Pharmaceuticals, Yongin-si 17028, Republic of Korea
| | - Gwan-Young Kim
- Life Science Research Institute, Daewoong Pharmaceuticals, Yongin-si 17028, Republic of Korea
| | - Min-Hwan Kim
- Life Science Research Institute, Daewoong Pharmaceuticals, Yongin-si 17028, Republic of Korea
| | | | - Min-Jae Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Mansingh Chaudhary
- Department of Biomedicine, Health & Life Convergence Sciences (BK21 Four) and Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Khadka Bikram
- Department of Biomedicine, Health & Life Convergence Sciences (BK21 Four) and Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Taeheon Kim
- Life Science Research Institute, Daewoong Pharmaceuticals, Yongin-si 17028, Republic of Korea
| | - Seungmok Choi
- Life Science Research Institute, Daewoong Pharmaceuticals, Yongin-si 17028, Republic of Korea
| | - Heejin Yang
- Life Science Research Institute, Daewoong Pharmaceuticals, Yongin-si 17028, Republic of Korea
| | - Joo Won Park
- Bio-Synectics, Inc., Seoul 08826, Republic of Korea
| | - Dae-Duk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Ki-Taek Kim
- Department of Biomedicine, Health & Life Convergence Sciences (BK21 Four) and Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| |
Collapse
|
21
|
Li Y, Zhang Z, Xie J, Lian X, Zhang G, Wang C. Bioequivalence Assessment of Two Dapoxetine Hydrochloride Formulations in Healthy Chinese Males Under Fasted and Fed Conditions. Clin Pharmacol Drug Dev 2024; 13:861-869. [PMID: 38396214 DOI: 10.1002/cpdd.1393] [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: 12/16/2023] [Accepted: 01/29/2024] [Indexed: 02/25/2024]
Abstract
This study evaluated the bioequivalence of the newly developed dapoxetine hydrochloride tablet relative to the marketed reference product by comparing their pharmacokinetic profiles under fasted and fed conditions. A total of 60 healthy Chinese male subjects participated in a single-center, 2-period, 2-sequence, randomized, open-label, self-crossover study with a washout period of 14 days, 30 in the fasted group and 30 in the fed group. Following a single 30-mg oral dose of the test or reference dapoxetine formulation, blood samples were collected before dosing to 72 hours after dosing. Liquid chromatography-tandem mass spectrometry was performed to measure plasma concentration of dapoxetine and determine pharmacokinetic parameters through noncompartmental analysis. The vital signs and adverse events were also monitored during the study. The 90% confidence intervals of the geometric mean ratios for maximum plasma concentration, area under the plasma concentration-time curve from time 0 to the last concentration time, and area under the plasma concentration-time curve from time 0 extrapolated to infinity of the 2 dapoxetine formulations completely fell within the regulatory criteria for bioequivalence of 80%-125%. In addition, both dapoxetine hydrochloride formulations were generally well tolerated. The generic dapoxetine hydrochloride tablet was bioequivalent to the marketed reference product in healthy Chinese men with no discernible safety differences.
Collapse
Affiliation(s)
- Yumin Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- School of Chemical Engineering, Xuzhou Vocational College of Industrial Technology, Xuzhou, China
| | - Zhen Zhang
- Shandong Hubble Kisen Biological Technology CO., Ltd., Jinan, China
| | - Jizhen Xie
- Shandong Center for Food and Drug Evaluation and Inspection, Jinan, China
| | - Xianghua Lian
- Shandong Hubble Kisen Biological Technology CO., Ltd., Jinan, China
| | - Guangtao Zhang
- Shandong Hubble Kisen Biological Technology CO., Ltd., Jinan, China
| | - Cheng Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
22
|
Antipas GSE, Reul R, Voges K, Kyeremateng SO, Ntallis NA, Karalis KT, Miroslaw L. System-agnostic prediction of pharmaceutical excipient miscibility via computing-as-a-service and experimental validation. Sci Rep 2024; 14:15106. [PMID: 38956156 PMCID: PMC11219749 DOI: 10.1038/s41598-024-65978-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024] Open
Abstract
We applied computing-as-a-service to the unattended system-agnostic miscibility prediction of the pharmaceutical surfactants, Vitamin E TPGS and Tween 80, with Copovidone VA64 polymer at temperature relevant for the pharmaceutical hot melt extrusion process. The computations were performed in lieu of running exhaustive hot melt extrusion experiments to identify surfactant-polymer miscibility limits. The computing scheme involved a massively parallelized architecture for molecular dynamics and free energy perturbation from which binodal, spinodal, and mechanical mixture critical points were detected on molar Gibbs free energy profiles at 180 °C. We established tight agreement between the computed stability (miscibility) limits of 9.0 and 10.0 wt% vs. the experimental 7 and 9 wt% for the Vitamin E TPGS and Tween 80 systems, respectively, and identified different destabilizing mechanisms applicable to each system. This paradigm supports that computational stability prediction may serve as a physically meaningful, resource-efficient, and operationally sensible digital twin to experimental screening tests of pharmaceutical systems. This approach is also relevant to amorphous solid dispersion drug delivery systems, as it can identify critical stability points of active pharmaceutical ingredient/excipient mixtures.
Collapse
Affiliation(s)
| | - Regina Reul
- AbbVie Deutschland GmbH & Co. KG, Development Sciences, 67061, Ludwigshafen, Germany
| | - Kristin Voges
- AbbVie Deutschland GmbH & Co. KG, Development Sciences, 67061, Ludwigshafen, Germany
| | - Samuel O Kyeremateng
- AbbVie Deutschland GmbH & Co. KG, Development Sciences, 67061, Ludwigshafen, Germany.
| | | | | | - Lukasz Miroslaw
- Azure High Performance Computing and Artificial Intelligence, Microsoft Switzerland, The Circle 02, 8058, Zurich, Switzerland
| |
Collapse
|
23
|
Neusaenger AL, Fatina C, Yu J, Yu L. Effect of Polymer Architecture and Acidic Group Density on the Degree of Salt Formation in Amorphous Solid Dispersions. Mol Pharm 2024; 21:3375-3382. [PMID: 38885189 DOI: 10.1021/acs.molpharmaceut.4c00089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Recent work has shown that an amorphous drug-polymer salt can be highly stable against crystallization under hot and humid storage conditions (e.g., 40 °C/75% RH) and provide fast release and that these advantages depend on the degree of salt formation. Here, we investigate the salt formation between the basic drug lumefantrine (LMF) and several acidic polymers: poly(acrylic acid) (PAA), hypromellose phthalate (HPMCP), hypromellose acetate succinate (HPMCAS), cellulose acetate phthalate (CAP), Eudragit L100, and Eudragit L100-55. Salt formation was performed by "slurry synthesis" where dry components were mixed at room temperature in the presence of a small quantity of an organic solvent, which was subsequently removed. This method achieved more complete salt formation than the conventional methods of hot-melt extrusion and rotary evaporation. The acidic group density of a polymer was determined by nonaqueous titration in the same solvent used for slurry synthesis; the degree of LMF protonation was determined by X-ray photoelectron spectroscopy. The polymers studied show very different abilities to protonate LMF when compared at a common drug loading, following the order PAA > (HPMCP ∼ CAP ∼ L100 ∼ L100-55) > HPMCAS, but the difference largely disappears when the degree of protonation is plotted against the concentration of the available acidic groups for reaction. This indicates that the extent of salt formation is mainly controlled by the acidic group density and is less sensitive to the polymer architecture. Our results are relevant for selecting the optimal polymer to control the degree of ionization in amorphous solid dispersions.
Collapse
Affiliation(s)
- Amy Lan Neusaenger
- School of Pharmacy, University of Wisconsin, 777 Highland Ave., Madison, Wisconsin 53705, United States
| | - Caroline Fatina
- School of Pharmacy, University of Wisconsin, 777 Highland Ave., Madison, Wisconsin 53705, United States
| | - Junguang Yu
- School of Pharmacy, University of Wisconsin, 777 Highland Ave., Madison, Wisconsin 53705, United States
| | - Lian Yu
- School of Pharmacy, University of Wisconsin, 777 Highland Ave., Madison, Wisconsin 53705, United States
- Department of Chemistry, University of Wisconsin, 1101 University Ave., Madison, Wisconsin 53706, United States
| |
Collapse
|
24
|
Li S, Abdelquader MM, Andrews GP, Jones DS. Towards a greater understanding of the deep eutectic phenomenon through examination of the lidocaine-NSAID therapeutic deep eutectic systems. Eur J Pharm Biopharm 2024; 200:114329. [PMID: 38761870 DOI: 10.1016/j.ejpb.2024.114329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/29/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
Therapeutic deep eutectic solvents (THEDES) have been attracting increasing attention in the pharmaceutical literature as a promising enabling technology capable of improving physicochemical and biopharmaceutical properties for difficult-to-deliver drug compounds. The current literature has explored amide local anaesthetics and carboxylic acid nonsteroidal anti-inflammatories (NSAIDs) as commonly used THEDES formers for their active hydrogen-bonding functionality. However, little is known about what happens within the "deep eutectic" region where a range of binary compositions present simply as a liquid with no melting events detectable across experimentally achievable conditions. There is also very limited understanding of how parent compounds' physicochemical properties could impact upon the formation, interaction mechanism, and stability of the formed liquid systems, despite the significance of these information in dose adjustment, industrial handling, and scaling-up of these liquids. In the current work, we probed the "deep eutectic" phenomenon by investigating the formation and physicochemical behaviours of some chosen lidocaine-NSAID systems across a wide range of composition ratios. Our data revealed that successfully formed THEDES exhibited composition dependent Tg variations with strong positive deviations from predicted Tg values using the Gordon-Taylor theory, suggesting substantial interactions within the formed supramolecular structure. Interestingly, it was found that the parent compound's glass forming ability had a noticeable impact upon such profound interaction and hence could dictate the success of THEDES formation. It has also been confirmed that all successful systems were formed based on charge-assisted hydrogen bonding within their THEDES network, affirming the significant role of partial protonisation on achieving a profound melting point depression. More importantly, the work found that within the "deep eutectic" region there was still an ideal, or thermodynamically preferrable "THEDES point", which would exhibit excellent stability upon exposure to stress storage conditions. The discoveries of this study bring the literature one step closer to fully understanding the "therapeutic deep eutectic" phenomenon. Through correlation between parent reagents' physicochemical properties and the synthesised products' characteristics, we establish a more educated process for the prediction and engineering of THEDES.
Collapse
Affiliation(s)
- Shu Li
- Pharmaceutical Engineering Group, School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK.
| | - Magdy M Abdelquader
- Pharmaceutical Engineering Group, School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; Pharmaceutical Technology Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| | - Gavin P Andrews
- Pharmaceutical Engineering Group, School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK.
| | - David S Jones
- Pharmaceutical Engineering Group, School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK.
| |
Collapse
|
25
|
Matchimabura N, Praparatana R, Issarachot O, Oungbho K, Wiwattanapatapee R. Development of raft-forming liquid formulations loaded with ginger extract-solid dispersion for treatment of gastric ulceration. Heliyon 2024; 10:e31803. [PMID: 38841494 PMCID: PMC11152664 DOI: 10.1016/j.heliyon.2024.e31803] [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: 12/06/2023] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024] Open
Abstract
Raft-forming liquid formulations incorporating ginger extract solid dispersion (GE-SD) were developed to achieve prolonged delivery of 6-gingerol in the stomach and thus increase the effectiveness of gastric ulcer treatment. The solubility of 6-gingerol in 0.1 N HCl (pH 1.2) was maximized (15 mg/mL) by combining ginger extract with PVP K30 at 1:3 w/w ratio to produce a solid dispersion. The nature of GE-SD was confirmed by PXRD and FT-IR analysis. PXRD pattern showed miscibility of GE and PVP K30 in amorphous solid dispersion and the FT-IR spectra confirmed the formation of hydrogen bond between GE and PVP K30. GE-SD-loaded raft-forming liquids were prepared using sodium alginate as a gel former and HPMC as a release-controlling agent. The formulations exhibited rapid floating behavior in 0.1 N HCl (<30 s) and remained afloat on the surface over 8 h. The formed raft structures provided sufficient strength (>7.5 g) and allowed sustained release of more than 70 % of the 6-gingerol content over 8 h in 0.1 N HCl. Raft-forming formulations incorporating ginger extract demonstrated anti-inflammatory activity by inhibiting nitric oxide production in LPS-stimulated RAW 264.7 macrophage cells (IC50 = 5.13 ± 0.07 μg/mL). Exposure to the formulations also had a significant cytotoxic effect on AGS human gastric adenocarcinoma cells with an IC50 of 17.45 ± 0.29 μg/mL. In addition, the raft-forming formulations enhanced the migratory behavior of L929 mouse fibroblasts in the scratch wound model. Taken together, these findings reveal the benefits of gastro-retentive, GE-SD-loaded raft-forming liquid formulations for improving the treatment of gastric ulcers.
Collapse
Affiliation(s)
- Nattawipa Matchimabura
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla, 90112, Thailand
| | - Rachanida Praparatana
- Faculty of Medical Technology, Prince of Songkla University, Hat Yai, Songkhla, 90112, Thailand
| | - Ousanee Issarachot
- Department of Pharmacy Technician, Sirindhorn College of Public Health Trang, Faculty of Public Health and Allied Health Sciences, Praboromarajchanok Institute, Trang, 92110, Thailand
| | - Kwunchit Oungbho
- Medical Science Research and Innovation, Prince of Songkla University, Hat Yai, Songkhla, 90112, Thailand
- Drug Delivery System Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla, 90112, Thailand
| | - Ruedeekorn Wiwattanapatapee
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla, 90112, Thailand
- Phytomedicine and Pharmaceutical Biotechnology Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hatyai, Songkhla, 90112, Thailand
| |
Collapse
|
26
|
Ali ISM, Sajad UA, Abdul Rasool BK. Solid dispersion systems for enhanced dissolution of poorly water-soluble candesartan cilexetil: In vitro evaluation and simulated pharmacokinetics studies. PLoS One 2024; 19:e0303900. [PMID: 38843120 PMCID: PMC11156308 DOI: 10.1371/journal.pone.0303900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/02/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Candesartan cilexetil (CC) is a selective angiotensin II receptor antagonist widely used to treat hypertension. CC is a substrate of P-glycoprotein (P-gp), causing its efflux to the intestinal lumen. It is also practically insoluble in water and has low oral bioavailability (14%). Thus, the current study aims to improve the in vitro dissolution of CC by developing solid dispersion systems (SDSs) and corroborating the in vitro results using a simulated pharmacokinetics study. METHODS The SDSs were prepared using polyvinyl pyrrolidone (PVP) as a water-soluble polymer, Eudragit E100 (EE100) as a pH-dependent soluble carrier, and a combination of these two polymers. The saturation solubility and the dissolution rate studies of the prepared systems in three dissolution media were performed. The optimized system SE-EE5 was selected for further investigations, including DSC, XRD, FTIR, FESEM, DLS, TSEM, IVIVC convolution study, and stability studies. RESULTS The solubility of CC significantly increased by a factor of 27,037.344 when formulated as a solid dispersion matrix using EE100 at a ratio of 1:5 (w/w) drug to polymer (SE-EE5 SD), compared to the solubility of the pure drug. The mechanism of solubility and dissolution rate enhancement of CC by the optimized SDS was found to be via the conversion of the crystalline CC into the amorphous form as well as nanoparticles formation upon dissolution at a pH below 5. The instrumental analysis tests showed good compatibility between CC and EE100 and there was no chemical interaction between the drug and the polymer. Moreover, the stability tests confirmed that the optimized system was stable after three months of storage at 25°C. CONCLUSION The utilization of the solid dispersion technique employing EE 100 polymer as a matrix demonstrates significant success in enhancing the solubility, dissolution, and subsequently, the bioavailability of water-insoluble drugs like CC.
Collapse
Affiliation(s)
| | - Uday Aziz Sajad
- Pharmaceutics Department, College of Pharmacy, University of Basrah, Basrah, Iraq
- Pharmaceutics Department, College of Pharmacy, Almaaqal University, Basrah, Iraq
| | - Bazigha K. Abdul Rasool
- Department of Pharmaceutical Sciences, Dubai Pharmacy College for Girls, Dubai, United Arab Emirates
| |
Collapse
|
27
|
Giannachi C, Allen E, Egan G, Vucen S, Crean A. Colyophilized Sugar-Polymer Dispersions for Enhanced Processing and Storage Stability. Mol Pharm 2024; 21:3017-3026. [PMID: 38758116 DOI: 10.1021/acs.molpharmaceut.4c00187] [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: 05/18/2024]
Abstract
Sucrose and trehalose pharmaceutical excipients are employed to stabilize protein therapeutics in a dried state. The mechanism of therapeutic protein stabilization is dependent on the sugars being present in an amorphous solid-state. Colyophilization of sugars with high glass transition polymers, polyvinylpyrrolidone (PVP), and poly(vinylpyrrolidone vinyl acetate) (PVPVA), enhances amorphous sugar stability. This study investigates the stability of colyophilized sugar-polymer systems in the frozen solution state, dried state postlyophilization, and upon exposure to elevated humidity. Binary systems of sucrose or trehalose with PVP or PVPVA were lyophilized with sugar/polymer ratios ranging from 2:8 to 8:2. Frozen sugar-PVPVA solutions exhibited a higher glass transition temperature of the maximally freeze-concentrated amorphous phase (Tg') compared to sugar-PVP solutions, despite the glass transition temperature (Tg) of PVPVA being lower than PVP. Tg values of all colyophilized systems were in a similar temperature range irrespective of polymer type. Greater hydrogen bonding between sugars and PVP and the lower hygroscopicity of PVPVA influenced polymer antiplasticization effects and the plasticization effects of residual water. Plasticization due to water sorption was investigated in a dynamic vapor sorption humidity ramping experiment. Lyophilized sucrose systems exhibited increased amorphous stability compared to trehalose upon exposure to the humidity. Recrystallization of trehalose was observed and stabilized by polymer addition. Lower concentrations of PVP inhibited trehalose recrystallization compared to PVPVA. These stabilizing effects were attributed to the increased hydrogen bonding between trehalose and PVP compared to trehalose and PVPVA. Overall, the study demonstrated how differences in polymer hygroscopicity and hydrogen bonding with sugars influence the stability of colyophilized amorphous dispersions. These insights into excipient solid-state stability are relevant to the development of stabilized biopharmaceutical solid-state formulations.
Collapse
Affiliation(s)
- Claudia Giannachi
- SSPC, the SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Cork T12 YT20, Ireland
- School of Pharmacy, University College Cork, Cork T12 YT20, Ireland
| | - Evin Allen
- School of Pharmacy, University College Cork, Cork T12 YT20, Ireland
| | - Gráinne Egan
- School of Pharmacy, University College Cork, Cork T12 YT20, Ireland
| | - Sonja Vucen
- SSPC, the SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Cork T12 YT20, Ireland
- School of Pharmacy, University College Cork, Cork T12 YT20, Ireland
| | - Abina Crean
- SSPC, the SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Cork T12 YT20, Ireland
- School of Pharmacy, University College Cork, Cork T12 YT20, Ireland
| |
Collapse
|
28
|
Chen Z, Gao W, Feng X, Zhou G, Zhang M, Zeng L, Hu X, Liu Z, Song H. A comparative study on the preparation and evaluation of solubilizing systems for silymarin. Drug Deliv Transl Res 2024; 14:1616-1634. [PMID: 37964172 DOI: 10.1007/s13346-023-01476-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2023] [Indexed: 11/16/2023]
Abstract
Silymarin (SM) exhibits clinical efficacy in treating liver injuries, cirrhosis, and chronic hepatitis. However, its limited water solubility and low bioavailability hinder its therapeutic potential. The primary objective of this study was to compare the in vitro and in vivo characteristics of the four distinct SM solubilization systems, namely SM solid dispersion (SM-SD), SM phospholipid complex (SM-PC), SM sulfobutyl ether-β-cyclodextrin inclusion complex (SM-SBE-β-CDIC) and SM self-microemulsifying drug delivery system (SM-SMEDDS) to provide further insights into their potential for enhancing the solubility and bioavailability of SM. The formation of SM-SD, SM-PC, and SM-SBE-β-CDIC was thoroughly characterized using scanning electron microscopy (SEM), Fourier transform infrared spectroscopy (FTIR), and powder X-ray diffractometry (PXRD) techniques to analyze the changes in their microscopic structure, molecular structure, and crystalline state. The particle size and polydispersity index (PDI) of SM-SMEDDS were 71.6 ± 1.57 nm, and 0.13 ± 0.03, respectively. The self-emulsifying time of SM-SMEDDS was 3.0 ± 0.3 min. SM-SMEDDS exhibited an improved in vitro dissolution rate and demonstrated the highest relative bioavailability compared to pure SM, SM-SD, SM-PC, SM-SBE-β-CDIC, and Legalon®. Consequently, SMEDDS shows promise as a drug delivery system for orally administered SM, offering enhanced solubility and bioavailability.
Collapse
Affiliation(s)
- Zhenzhen Chen
- Department of Pharmacy, Fuzong Clinical Medical College of Fujian Medical University (900 Hospital of the Joint Logistics Team), Fuzhou, 350025, PR China
| | - Wenhao Gao
- Department of Pharmacy, Fuzong Clinical Medical College of Fujian Medical University (900 Hospital of the Joint Logistics Team), Fuzhou, 350025, PR China
| | - Xianquan Feng
- Department of Pharmacy, Fuzong Clinical Medical College of Fujian Medical University (900 Hospital of the Joint Logistics Team), Fuzhou, 350025, PR China
| | - Guizhi Zhou
- Department of Pharmacy, Fuzong Clinical Medical College of Fujian Medical University (900 Hospital of the Joint Logistics Team), Fuzhou, 350025, PR China
- School of Pharmacy, Fujian University of Chinese Traditional Medicine, Fuzhou, 350108, China
| | - Minxin Zhang
- Department of Pharmacy, Fuzong Clinical Medical College of Fujian Medical University (900 Hospital of the Joint Logistics Team), Fuzhou, 350025, PR China
| | - Lingjun Zeng
- Department of Pharmacy, Fuzong Clinical Medical College of Fujian Medical University (900 Hospital of the Joint Logistics Team), Fuzhou, 350025, PR China
| | - Xiaomu Hu
- Department of Pharmacy, Fuzong Clinical Medical College of Fujian Medical University (900 Hospital of the Joint Logistics Team), Fuzhou, 350025, PR China
| | - Zhihong Liu
- Department of Pharmacy, Fuzong Clinical Medical College of Fujian Medical University (900 Hospital of the Joint Logistics Team), Fuzhou, 350025, PR China.
| | - Hongtao Song
- Department of Pharmacy, Fuzong Clinical Medical College of Fujian Medical University (900 Hospital of the Joint Logistics Team), Fuzhou, 350025, PR China.
| |
Collapse
|
29
|
Kokott M, Breitkreutz J, Wiedey R. The interplay of poorly soluble drugs in dissolution from amorphous solid dispersions. Int J Pharm X 2024; 7:100243. [PMID: 38585343 PMCID: PMC10997824 DOI: 10.1016/j.ijpx.2024.100243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/09/2024] Open
Abstract
In recent years, the application of fixed dose combinations of antiretroviral drugs in HIV therapy has been established. Despite numerous therapeutic benefits, this approach poses several challenges for the formulation development especially when poorly soluble drugs are considered. Amorphous solid dispersions (ASD) thereby have gained considerable interest in the pharmaceutical field, however, mainly including binary systems containing only one drug and a polymer. The co-formulation of two amorphous drugs can be accompanied by an immense increase in the complexity of the system as exemplarily reported for ritonavir and lopinavir embedded in a composite polymer matrix of PVPVA. The present study aims to present a new formulation approach to overcome the well-documented interaction during dissolution. Two different polymers, PVPVA and HPMCAS were used to produce ASDs for both drugs individually via hot-melt extrusion. The embedding of lopinavir in the slower dissolving polymer HPMCAS, while using PVPVA for ritonavir was found to significantly improve the overall dissolution performance compared to the individual use of PVPVA as well as to the commercial product Kaletra®. In addition, the use of different grades of HPMCAS demonstrated the possibility to further modify the dissolution profile. For a preliminary biorelevant assessment, the selected formulations were tested in a biphasic dissolution setup.
Collapse
Affiliation(s)
- Marcel Kokott
- Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University, Duesseldorf Universitaetsstr. 1, Duesseldorf 40225, Germany
| | - Jörg Breitkreutz
- Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University, Duesseldorf Universitaetsstr. 1, Duesseldorf 40225, Germany
| | - Raphael Wiedey
- Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University, Duesseldorf Universitaetsstr. 1, Duesseldorf 40225, Germany
| |
Collapse
|
30
|
Yusuf H, Savitri OMN, Al-Khalifi NN, Gunawan L, Chairul BK, Rijal MAS, Isadiartuti D, Sari R. Cellulose- and Saccharide-Based Orally Dispersible Thin Films Transform the Solid States and Dissolution Characteristics of Poorly Soluble Curcumin. Adv Pharmacol Pharm Sci 2024; 2024:8596712. [PMID: 38831896 PMCID: PMC11147675 DOI: 10.1155/2024/8596712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 04/08/2024] [Accepted: 05/06/2024] [Indexed: 06/05/2024] Open
Abstract
This study aimed at developing and optimizing the orally dispersible thin film (ODTF) containing a plant-derived drug-curcumin (CUR). CUR belongs to a biopharmaceutical classification system (BCS) class IV compound that requires improving its water solubility and tissue permeability preceding formulation. An ODTF was applied to produce a solid dispersion matrix for CUR to resolve such solubility and permeability problems. The film-forming polymers used in the study were cellulose-based (hydroxypropyl methylcellulose/HPMC and carboxymethylcellulose/CMC) and saccharide-based maltodextrin (MDX). Poloxamer (POL) was also employed as surfactant and solubilizer. The solvent casting technique was applied to produce the films. The ethanolic solution of CUR was mixed with an aqueous solution of POLs and then incorporated into different film-forming polymers prior to casting. The processing of the CUR with POL solution was intended to aid in the even dispersion of the drug in the polymeric matrices and enhance the wettability of the films. The physical state and properties of the films were characterized in terms of their morphology, crystallinity of the drug, and phase miscibility of the mixtures. The dissolution profile of the films was also evaluated in terms of dissolution rate and dissolution efficiency. The obtained ODTF products were smooth and flat-surfaced. Physical characterization also indicated that the CUR was homogeneously dispersed in the ODTFs and no longer existed as crystalline material as revealed by X-ray diffraction (XRD). The CUR was also not phase-separated from the films as disclosed by differential scanning calorimetry (DSC). Such dispersion was achieved through the solubilizing effect of POLs and compact polymeric film matrices that prevented the CUR from recrystallization. Furthermore, the ODTFs also improved the dissolution of CUR by 3.2-fold higher than the raw CUR. Overall, cellulose-based films had favorable physical properties compared with saccharide-based films.
Collapse
Affiliation(s)
- Helmy Yusuf
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Jl. Mulyorejo, Surabaya 60115, Indonesia
| | | | - Nadia Natsya Al-Khalifi
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Jl. Mulyorejo, Surabaya 60115, Indonesia
| | - Lavinia Gunawan
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Jl. Mulyorejo, Surabaya 60115, Indonesia
| | - Brian Karno Chairul
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Jl. Mulyorejo, Surabaya 60115, Indonesia
| | - M. Agus Syamsur Rijal
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Jl. Mulyorejo, Surabaya 60115, Indonesia
| | - Dewi Isadiartuti
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Jl. Mulyorejo, Surabaya 60115, Indonesia
| | - Retno Sari
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Jl. Mulyorejo, Surabaya 60115, Indonesia
| |
Collapse
|
31
|
Di R, Bansal KK, Rosenholm JM, Grohganz H, Rades T. Utilizing the allyl-terminated copolymer methoxy(poly(ethylene glycol))-block-poly(jasmine lactone) in the development of amorphous solid dispersions: A comparative study of functionalized and non-functionalized polymer. Int J Pharm 2024; 657:124175. [PMID: 38685442 DOI: 10.1016/j.ijpharm.2024.124175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
Molecular interactions are crucial to stabilize amorphous drugs in amorphous solid dispersions (ASDs). Most polymers, however, have only a limited ability to form strong molecular interactions with drugs. Polymers tailored to fit the physicochemical properties of the drug molecule to be incorporated, for instance by allowing the incorporation of specific functional groups, would be highly sought-for in this regard. For this purpose, the novel allyl-terminated polymer methoxy(polyethylene glycol)-block-poly(jasmine lactone) (mPEG-b-PJL) has been synthesized and functionalized to potentially enhance specific drug-polymer interactions. This study investigated the use of mPEG-b-PJL in ASDs, using carvedilol (CAR), a weakly basic model drug. The findings revealed that the acidic functionalized form of the polymer (mPEG-b-PJL-COOH) indeed established stronger molecular interactions with CAR compared to its non-functionalized counterpart mPEG-b-PJL. Evaluations on polymer effectiveness in forming ASDs demonstrated that mPEG-b-PJL-COOH outperformed its non-functionalized counterpart in miscibility, drug loading ability, and stability, inferred from reduced molecular mobility. However, dissolution tests indicated that ASDs with mPEG-b-PJL-COOH did not significantly improve the dissolution behaviour compared to amorphous CAR alone, despite potential solubility enhancement through micelle formation. Overall, this study confirms the potential of functionalized polymers in ASD formulations, while the challenge of improving dissolution performance in these ASDs remains an area of further development.
Collapse
Affiliation(s)
- Rong Di
- University of Copenhagen, Faculty of Health and Medical Sciences, Department of Pharmacy, Copenhagen, Denmark.
| | - Kuldeep K Bansal
- Åbo Akademi University, Faculty of Science and Engineering, Pharmaceutical Sciences Laboratory, Turku, Finland.
| | - Jessica M Rosenholm
- Åbo Akademi University, Faculty of Science and Engineering, Pharmaceutical Sciences Laboratory, Turku, Finland.
| | - Holger Grohganz
- University of Copenhagen, Faculty of Health and Medical Sciences, Department of Pharmacy, Copenhagen, Denmark.
| | - Thomas Rades
- University of Copenhagen, Faculty of Health and Medical Sciences, Department of Pharmacy, Copenhagen, Denmark.
| |
Collapse
|
32
|
Liu Y, Liang Y, Yuhong J, Xin P, Han JL, Du Y, Yu X, Zhu R, Zhang M, Chen W, Ma Y. Advances in Nanotechnology for Enhancing the Solubility and Bioavailability of Poorly Soluble Drugs. Drug Des Devel Ther 2024; 18:1469-1495. [PMID: 38707615 PMCID: PMC11070169 DOI: 10.2147/dddt.s447496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/03/2024] [Indexed: 05/07/2024] Open
Abstract
This manuscript offers a comprehensive overview of nanotechnology's impact on the solubility and bioavailability of poorly soluble drugs, with a focus on BCS Class II and IV drugs. We explore various nanoscale drug delivery systems (NDDSs), including lipid-based, polymer-based, nanoemulsions, nanogels, and inorganic carriers. These systems offer improved drug efficacy, targeting, and reduced side effects. Emphasizing the crucial role of nanoparticle size and surface modifications, the review discusses the advancements in NDDSs for enhanced therapeutic outcomes. Challenges such as production cost and safety are acknowledged, yet the potential of NDDSs in transforming drug delivery methods is highlighted. This contribution underscores the importance of nanotechnology in pharmaceutical engineering, suggesting it as a significant advancement for medical applications and patient care.
Collapse
Affiliation(s)
- Yifan Liu
- School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Yushan Liang
- School of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Jing Yuhong
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Peng Xin
- School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Jia Li Han
- School of Health Sciences, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Yongle Du
- School of Ophthalmology and Optometry, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Xinru Yu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Runhe Zhu
- School of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Mingxun Zhang
- School of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Wen Chen
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| | - Yingjie Ma
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
33
|
Campos Pacheco JE, Yalovenko T, Riaz A, Kotov N, Davids C, Persson A, Falkman P, Feiler A, Godaly G, Johnson CM, Ekström M, Pilkington GA, Valetti S. Inhalable porous particles as dual micro-nano carriers demonstrating efficient lung drug delivery for treatment of tuberculosis. J Control Release 2024; 369:231-250. [PMID: 38479444 DOI: 10.1016/j.jconrel.2024.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 05/24/2024]
Abstract
Inhalation therapy treating severe infectious disease is among the more complex and emerging topics in controlled drug release. Micron-sized carriers are needed to deposit drugs into the lower airways, while nano-sized carriers are of preference for cell targeting. Here, we present a novel and versatile strategy using micron-sized spherical particles with an excellent aerodynamic profile that dissolve in the lung fluid to ultimately generate nanoparticles enabling to enhance both extra- and intra-cellular drug delivery (i.e., dual micro-nano inhalation strategy). The spherical particles are synthesised through the condensation of nano-sized amorphous silicon dioxide resulting in high surface area, disordered mesoporous silica particles (MSPs) with monodispersed size of 2.43 μm. Clofazimine (CLZ), a drug shown to be effective against multidrug-resistant tuberculosis, was encapsulated in the MSPs obtaining a dry powder formulation with high respirable fraction (F.P.F. <5 μm of 50%) without the need of additional excipients. DSC, XRPD, and Nitrogen adsorption-desorption indicate that the drug was fully amorphous when confined in the nano-sized pores (9-10 nm) of the MSPs (shelf-life of 20 months at 4 °C). Once deposited in the lung, the CLZ-MSPs exhibited a dual action. Firstly, the nanoconfinement within the MSPs enabled a drastic dissolution enhancement of CLZ in simulated lung fluid (i.e., 16-fold higher than the free drug), increasing mycobacterial killing than CLZ alone (p = 0.0262) and reaching concentrations above the minimum bactericidal concentration (MBC) against biofilms of M. tuberculosis (i.e., targeting extracellular bacteria). The released CLZ permeated but was highly retained in a Calu-3 respiratory epithelium model, suggesting a high local drug concentration within the lung tissue minimizing risk for systemic side effects. Secondly, the micron-sized drug carriers spontaneously dissolve in simulated lung fluid into nano-sized drug carriers (shown by Nano-FTIR), delivering high CLZ cargo inside macrophages and drastically decreasing the mycobacterial burden inside macrophages (i.e., targeting intracellular bacteria). Safety studies showed neither measurable toxicity on macrophages nor Calu-3 cells, nor impaired epithelial integrity. The dissolved MSPs also did not show haemolytic effect on human erythrocytes. In a nutshell, this study presents a low-cost, stable and non-invasive dried powder formulation based on a dual micro-nano carrier to efficiently deliver drug to the lungs overcoming technological and practical challenges for global healthcare.
Collapse
Affiliation(s)
- Jesús E Campos Pacheco
- Biomedical Science, Faculty of Health and Society, Malmö University, 205 06 Malmö, Sweden; Biofilms - Research Center for Biointerfaces (BRCB), Malmö University, 205 06 Malmö, Sweden
| | - Tetiana Yalovenko
- Biomedical Science, Faculty of Health and Society, Malmö University, 205 06 Malmö, Sweden; Biofilms - Research Center for Biointerfaces (BRCB), Malmö University, 205 06 Malmö, Sweden
| | - Azra Riaz
- Biomedical Science, Faculty of Health and Society, Malmö University, 205 06 Malmö, Sweden; Biofilms - Research Center for Biointerfaces (BRCB), Malmö University, 205 06 Malmö, Sweden
| | - Nikolay Kotov
- Department of Chemistry, KTH Royal Institute of Technology, SE-100 44 Stockholm, Sweden
| | - Camilla Davids
- Department of Microbiology, Immunology and Glycobiology, Institution of Laboratory Medicine, Lund University, Lund, Sweden
| | - Alva Persson
- Biomedical Science, Faculty of Health and Society, Malmö University, 205 06 Malmö, Sweden; Biofilms - Research Center for Biointerfaces (BRCB), Malmö University, 205 06 Malmö, Sweden
| | - Peter Falkman
- Biomedical Science, Faculty of Health and Society, Malmö University, 205 06 Malmö, Sweden; Biofilms - Research Center for Biointerfaces (BRCB), Malmö University, 205 06 Malmö, Sweden
| | - Adam Feiler
- Department of Chemistry, KTH Royal Institute of Technology, SE-100 44 Stockholm, Sweden; Nanologica AB (publ), Forskargatan 20G, 151 36 Södertälje, Sweden
| | - Gabriela Godaly
- Department of Microbiology, Immunology and Glycobiology, Institution of Laboratory Medicine, Lund University, Lund, Sweden
| | - C Magnus Johnson
- Department of Chemistry, KTH Royal Institute of Technology, SE-100 44 Stockholm, Sweden
| | | | - Georgia A Pilkington
- Department of Chemistry, KTH Royal Institute of Technology, SE-100 44 Stockholm, Sweden; Nanologica AB (publ), Forskargatan 20G, 151 36 Södertälje, Sweden.
| | - Sabrina Valetti
- Biomedical Science, Faculty of Health and Society, Malmö University, 205 06 Malmö, Sweden; Biofilms - Research Center for Biointerfaces (BRCB), Malmö University, 205 06 Malmö, Sweden.
| |
Collapse
|
34
|
Autzen Virtanen A, Myślińska M, Healy AM, Power E, Madi A, Sivén M. The challenge of downstream processing of spray dried amorphous solid dispersions into minitablets designed for the paediatric population - A sustainable product development approach. Eur J Pharm Sci 2024; 196:106752. [PMID: 38518998 DOI: 10.1016/j.ejps.2024.106752] [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: 10/17/2023] [Revised: 03/08/2024] [Accepted: 03/19/2024] [Indexed: 03/24/2024]
Abstract
Poorly water-soluble drugs present a significant challenge in the development of oral solid dosage forms (OSDs). In formulation development the appropriate use of excipients to adjust solubility, and the choice of manufacturing method and pharmaceutical processes to obtain a dosage form to meet the needs of the patient group, is crucial. Preparing an amorphous solid dispersion (ASD) is a well-established method for solubility enhancement, and spray drying (SD) a common manufacturing method. However, the poor flowability of spray dried materials poses a significant challenge for downstream processing. Promoting sustainability in OSD development involves embracing a versatile formulation design, which enables a broader spectrum of patients to use the product, as opposed to altering existing dosage forms retrospectively. The objective of the current study was to develop a formulation of spray dried indomethacin ASD suited to the production, by direct compression, of instant release paediatric minitablets. Excipients evaluated were PVP or HPMCAS in solid dispersions at the preformulation phase, and MCC and lactose as a filler in direct compression. From the studied formulations, a 3:1 ratio blend of Vivapur 200/Pharmatose 200 M (MCC/lactose) with 0.5% (w/w) magnesium stearate was found to be the most promising in tableting, and minitablets containing a 6.22% content of spray-dried ASD of indomethacin/PVP K 29-32 could be obtained with desired tablet hardness and pharmaceutical quality, complying with tests of weight variation and fast disintegration in an aqueous environment. As a case example, this study provides a good foundation for further studies in harnessing a sustainable approach to the development of pharmaceutical formulations that can appropriately serve different patient sub-populations.
Collapse
Affiliation(s)
- Anja Autzen Virtanen
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Finland.
| | - Monika Myślińska
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland; SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals, Ireland
| | - Anne Marie Healy
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland; SSPC, the Science Foundation Ireland Research Centre for Pharmaceuticals, Ireland
| | - Eoin Power
- SK biotek Ireland, an SK pharmteco company, Ireland
| | - Atif Madi
- SK biotek Ireland, an SK pharmteco company, Ireland
| | - Mia Sivén
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Finland; Helsinki Institute of Sustainability Science, HELSUS, Finland
| |
Collapse
|
35
|
Sokač K, Miloloža M, Kučić Grgić D, Žižek K. Polymeric Amorphous Solid Dispersions of Dasatinib: Formulation and Ecotoxicological Assessment. Pharmaceutics 2024; 16:551. [PMID: 38675212 PMCID: PMC11053848 DOI: 10.3390/pharmaceutics16040551] [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: 02/26/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Dasatinib (DAS), a potent anticancer drug, has been subjected to formulation enhancements due to challenges such as significant first-pass metabolism, poor absorption, and limited oral bioavailability. To improve its release profile, DAS was embedded in a matrix of the hydrophilic polymer polyvinylpyrrolidone (PVP). Drug amorphization was induced in a planetary ball mill by solvent-free co-grinding, facilitating mechanochemical activation. This process resulted in the formation of amorphous solid dispersions (ASDs). The ASD capsules exhibited a notable enhancement in the release rate of DAS compared to capsules containing the initial drug. Given that anticancer drugs often undergo limited metabolism in the body with unchanged excretion, the ecotoxicological effect of the native form of DAS was investigated as well, considering its potential accumulation in the environment. The highest ecotoxicological effect was observed on the bacteria Vibrio fischeri, while other test organisms (bacteria Pseudomonas putida, microalgae Chlorella sp., and duckweed Lemna minor) exhibited negligible effects. The enhanced drug release not only contributes to improved oral absorption but also has the potential to reduce the proportion of DAS that enters the environment through human excretion. This comprehensive approach highlights the significance of integrating advances in drug development while considering its environmental implications.
Collapse
Affiliation(s)
- Katarina Sokač
- University of Zagreb, Faculty of Chemical Engineering and Technology, Trg Marka Marulića 19, 10000 Zagreb, Croatia; (D.K.G.); (K.Ž.)
| | - Martina Miloloža
- University of Zagreb, Faculty of Chemical Engineering and Technology, Trg Marka Marulića 19, 10000 Zagreb, Croatia; (D.K.G.); (K.Ž.)
| | | | | |
Collapse
|
36
|
Barr KE, Ohnsorg ML, Liberman L, Corcoran LG, Sarode A, Nagapudi K, Feder CR, Bates FS, Reineke TM. Drug-Polymer Nanodroplet Formation and Morphology Drive Solubility Enhancement of GDC-0810. Bioconjug Chem 2024; 35:499-516. [PMID: 38546823 DOI: 10.1021/acs.bioconjchem.4c00018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2024]
Abstract
Nanodroplet formation is important to achieve supersaturation of active pharmaceutical ingredients (APIs) in an amorphous solid dispersion. The aim of the current study was to explore how polymer composition, architecture, molar mass, and surfactant concentration affect polymer-drug nanodroplet morphology with the breast cancer API, GDC-0810. The impact of nanodroplet size and morphology on dissolution efficacy and drug loading capacity was explored using polarized light microscopy, dynamic light scattering, and cryogenic transmission electron microscopy. Poly(N-isopropylacrylamide-stat-N,N-dimethylacrylamide) (PND) was synthesized as two linear derivatives and two bottlebrush derivatives with carboxylated or PEGylated end-groups. Hydroxypropyl methylcellulose acetate succinate grade MF (HPMCAS-MF) and poly(vinylpyrrolidone-co-vinyl acetate) (PVPVA) were included as commercial polymer controls. We report the first copolymerization synthesis of a PVPVA bottlebrush copolymer, which was the highest performing excipient in this study, maintaining 688 μg/mL GDC-0810 concentration at 60 wt % drug loading. This is likely due to strong polymer-drug noncovalent interactions and the compaction of GDC-0810 along the PVPVA bottlebrush backbone. Overall, it was observed that the most effective formulations had a hydrodynamic radius less than 25 nm with tightly compacted nanodroplet morphologies.
Collapse
Affiliation(s)
- Kaylee E Barr
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Monica L Ohnsorg
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Lucy Liberman
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Louis G Corcoran
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Apoorva Sarode
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, California 94080, United States
| | - Karthik Nagapudi
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, California 94080, United States
| | - Christina R Feder
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, California 94080, United States
| | - Frank S Bates
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
37
|
Li J, Yu D, Zeng C, Mosquera-Giraldo LI, Everlof G, Foster K, Gesenberg C. Hydrolysis of Cellulose Acetate Phthalate and Hydroxypropyl Methylcellulose Phthalate in Amorphous Solid Dispersions. J Pharm Sci 2024:S0022-3549(24)00133-3. [PMID: 38608727 DOI: 10.1016/j.xphs.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024]
Abstract
The preparation of amorphous solid dispersions (ASDs) represents a promising strategy for addressing the solubility limitations of poorly soluble drugs, facilitating enhanced oral absorption. Acidic polymers such as cellulose acetate phthalate (CAP) and hydroxypropyl methylcellulose phthalate (HPMCP) have emerged as effective carriers for ASDs. Although the hydrolytic degradation of these polymers has been documented, its impact on the stability of ASDs has not been systematically investigated. This research aimed to explore the potential hydrolysis of CAP and HPMCP and how it influences the stability of ASDs containing ketoconazole (KTZ), at drug loadings of 10 % and 50 %. Our study utilized thermal analysis, infrared spectroscopy, and evaluations of physical and chemical stability. The results revealed that although KTZ remained physically stable in all ASDs over 60 days under various stability conditions, the emergence of crystalline phthalic acid (PA), a byproduct of polymer hydrolysis, was observed at elevated temperatures and relative humidity levels. The acidic microenvironment fostered by the release of PA further catalyzed drug chemical degradation. This study underscores the susceptibility of CAP and HPMCP to hydrolytic degradation, highlighting the inherent risk of PA-induced drug degradation, particularly for acid-labile compounds. These insights into the understanding of polymer hydrolysis in ASDs pave the way for the development of targeted approaches to safeguard drug stability and optimize pharmaceutical formulations for enhanced bioavailability, efficacy, and safety.
Collapse
Affiliation(s)
- Jinghan Li
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Dongyue Yu
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, NJ 08540, USA.
| | - Chaowang Zeng
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Laura I Mosquera-Giraldo
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, NJ 08540, USA
| | - Gerry Everlof
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, NJ 08540, USA
| | - Kimberly Foster
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, NJ 08540, USA
| | - Christoph Gesenberg
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, NJ 08540, USA
| |
Collapse
|
38
|
Benedikt Brenner M, Wüst M, Kuentz M, Wagner KG. High loading of lipophilic compounds in mesoporous silica for improved solubility and dissolution performance. Int J Pharm 2024; 654:123946. [PMID: 38417728 DOI: 10.1016/j.ijpharm.2024.123946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/22/2024] [Accepted: 02/22/2024] [Indexed: 03/01/2024]
Abstract
Loading poorly soluble active pharmaceutical ingredients (API) into mesoporous silica can enable API stabilization in non-crystalline form, which leads to improved dissolution. This is particularly beneficial for highly lipophilic APIs (log D7.4 > 8) as these drugs often exhibit limited solubility in dispersion forming carrier polymers, resulting in low drug load and reduced solid state stability. To overcome this challenge, we loaded the highly lipophilic natural products coenzyme Q10 (CoQ10) and astaxanthin (ASX), as well as the synthetic APIs probucol (PB) and lumefantrine (LU) into the mesoporous silica carriers Syloid® XDP 3050 and Silsol® 6035. All formulations were physically stable in their non-crystalline form and drug loads of up to 50 % were achieved. At increasing drug loads, a marked increase in equilibrium solubility of the active ingredients in biorelevant medium was detected, leading to improved performance during biorelevant biphasic dissolution studies (BiPHa + ). Particularly the natural products CoQ10 and ASX showed substantial benefits from being loaded into mesoporous carrier particles and clearly outperformed currently available commercial formulations. Performance differences between the model compounds could be explained by in silico calculations of the mixing enthalpy for drug and silica in combination with an experimental chromatographic method to estimate molecular interactions.
Collapse
Affiliation(s)
- Marvin Benedikt Brenner
- University of Bonn, Pharmaceutical Institute, Department of Pharmaceutics, Gerhard-Domagk-Str. 3, 53121 Bonn, Germany
| | - Matthias Wüst
- University of Bonn, Institute of Nutritional and Food Sciences, Food Chemistry, Friedrich-Hirzebruch-Allee 7, 53115 Bonn, Germany
| | - Martin Kuentz
- University of Applied Sciences and Arts Northwestern Switzerland, Institute of Pharma Technology, Hofackerstr. 30, 4132 Muttenz, Switzerland
| | - Karl G Wagner
- University of Bonn, Pharmaceutical Institute, Department of Pharmaceutics, Gerhard-Domagk-Str. 3, 53121 Bonn, Germany.
| |
Collapse
|
39
|
Danda LJDA, Amaral ARDC, Soares-Sobrinho JL, Soares MFDLR. Optimizing Nonsink Dissolution Testing for Amorphous Solid Dispersions: Exploring Sample Handling Variables. Mol Pharm 2024; 21:1861-1871. [PMID: 38416030 DOI: 10.1021/acs.molpharmaceut.3c01164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
This study aims to investigate key variables affecting the dissolution of amorphous pharmaceuticals. We examined sample treatment methods (centrifugation vs syringe filtration), time delays between sample collection and processing (immediate, 2, or 24 h), and different sample preparations (bare powder, capsules, or tablets). These factors were evaluated through both sink and nonsink dissolution experiments, using controlled supersaturation conditions (sink index ≈ 0.1) with amorphous solid dispersions (ASDs) containing low-substituted hydroxypropyl cellulose (L-HPC) and either indomethacin or posaconazole as model drugs. Our results highlighted the significant impact of syringe filtration on nonsink dissolutions, particularly the notable reduction in dissolved drug concentration, possibly due to filtration-induced precipitation. Moreover, introducing a delay of 2 or 24 h between sample collection and quantitation under nonsink conditions led to substantial concentration changes. This effect was not as pronounced when samples underwent centrifugation, and only the analysis was delayed for 2 h. The findings also emphasize the importance of accounting for delays introduced by pharmaceutical formulations, particularly in assessing the kinetic-solubility profiles of ASDs. This research offers valuable insights into the field of ASDs, enhancing our understanding of how these variables can influence dissolution results.
Collapse
|
40
|
Torabi A, Madsen FB, Skov AL. Permeation-Enhancing Strategies for Transdermal Delivery of Cannabinoids. Cannabis Cannabinoid Res 2024; 9:449-463. [PMID: 37751171 DOI: 10.1089/can.2023.0130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023] Open
Abstract
Introduction: This review aims to provide an overview of the advancements and status of clinical studies and potential permeation-enhancing strategies in the transdermal delivery of cannabinoids. Methods: A systematic and comprehensive literature search across academic databases, search engines, and online sources to identify relevant literature on the transdermal administration of cannabinoids. Results: Cannabinoids have proven beneficial in the treatment of wide-ranging physical and psychological disorders. A shift toward legalized cannabinoid products has increased both interests in cannabinoid research and the development of novel medicinal exploitations of cannabinoids in recent years. Oral and pulmonary delivery of cannabinoids has several limitations, including poor bioavailability, low solubility, and potential side effects. This has diverted scientific attention toward the transdermal route, successfully overcoming these hurdles by providing higher bioavailability, safety, and patient compliance. Yet, due to the barrier properties of the skin and the lipophilic nature of cannabinoids, there is a need to increase the permeation of the drugs to the underneath layers of skin to reach desired therapeutic plasma levels. Literature describing detailed clinical trials on cannabinoid transdermal delivery, either with or without permeation-enhancing strategies, is limited. Conclusion: The limited number of reports indicates that increased attention is needed on developing and examining efficient transdermal delivery systems for cannabinoids, including patch design and composition, drug-patch interaction, clinical effectiveness and safety in vivo, and permeation-enhancing strategies.
Collapse
Affiliation(s)
- Atefeh Torabi
- Danish Polymer Centre, Department of Chemical and Biochemical Engineering, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Frederikke Bahrt Madsen
- Danish Polymer Centre, Department of Chemical and Biochemical Engineering, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Anne Ladegaard Skov
- Danish Polymer Centre, Department of Chemical and Biochemical Engineering, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
41
|
Nagano K, Nakao T, Takeda M, Hirai H, Maekita H, Nakamura M, Imakawa N, Egawa A, Fujiwara T, Gao JQ, Kinoshita K, Sakata M, Nishino M, Yamashita T, Yoshida T, Harada K, Tachibana K, Doi T, Hirata K, Tsujino H, Higashisaka K, Tsutsumi Y. Polyglycerol fatty acid ester contributes to the improvement and maintenance of water solubility of amorphous curcumin by suppressing the intermolecular interaction and the diffusion rate of curcumin. Food Chem 2024; 437:137866. [PMID: 37931447 DOI: 10.1016/j.foodchem.2023.137866] [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: 11/06/2022] [Revised: 12/15/2022] [Accepted: 10/24/2023] [Indexed: 11/08/2023]
Abstract
Curcumin (CUR), a polyphenol, is an attractive component of functional foods, owing to various physiological activities. However, CUR is highly hydrophobic, insoluble in water, and difficult to absorb in the body. Here, we report an amorphous CUR formulation containing the dispersant polyglycerol fatty acid ester (PGFE), demonstrating high and stable water solubility. Improved water solubility enhanced the absorbability of CUR in our amorphous formulation along with enhanced triglyceride inhibition, compared to that in a commercial formulation. Nuclear Overhauser effect spectroscopy (NOESY) analysis revealed that PGFE reduced CUR-CUR interaction, resulting in higher dispersion and improved solubility of CUR. Taylor dispersion analysis showed a lower diffusion coefficient of CUR in the highly water-soluble formulation (with PGFE) than that in the low water-soluble formulation (without PGFE), which prevents recontact and recrystallization of CUR, which is trapped by PGFE. Overall, the amorphous CUR with high solubility could be used as a promising functional food.
Collapse
Affiliation(s)
- Kazuya Nagano
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; School of Pharmaceutical Sciences, Wakayama Medical University, 25-1 Shichibancho, Wakayama, Wakayama 640-8156, Japan.
| | - Tomohiro Nakao
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; San-Ei Gen F. F. I., Inc, 1-1-11 Sanwa-cho, Toyonaka, Osaka 561-8588, Japan
| | - Mariko Takeda
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Haruna Hirai
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hikaru Maekita
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Michiko Nakamura
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Naoki Imakawa
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ayako Egawa
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshimichi Fujiwara
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Jian-Qing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, PR China
| | - Keigo Kinoshita
- San-Ei Gen F. F. I., Inc, 1-1-11 Sanwa-cho, Toyonaka, Osaka 561-8588, Japan
| | - Makoto Sakata
- San-Ei Gen F. F. I., Inc, 1-1-11 Sanwa-cho, Toyonaka, Osaka 561-8588, Japan
| | - Masayuki Nishino
- San-Ei Gen F. F. I., Inc, 1-1-11 Sanwa-cho, Toyonaka, Osaka 561-8588, Japan
| | - Takuya Yamashita
- School of Pharmaceutical Sciences, Wakayama Medical University, 25-1 Shichibancho, Wakayama, Wakayama 640-8156, Japan
| | - Takuya Yoshida
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kazuo Harada
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Keisuke Tachibana
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takefumi Doi
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kazumasa Hirata
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; School of Pharmaceutical Sciences, Wakayama Medical University, 25-1 Shichibancho, Wakayama, Wakayama 640-8156, Japan
| | - Hirofumi Tsujino
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; The Museum of Osaka University, 1-13 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Kazuma Higashisaka
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Institute for Advanced Co-Creation Studies, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yasuo Tsutsumi
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, 1-6, Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
42
|
Zhuo X, Tozzetti M, Arnous A, Leng D, Foderà V, Löbmann K. Investigating the influence of protein secondary structure on the dissolution behavior of β-lactoglobulin-based amorphous solid dispersions. Int J Pharm 2024; 653:123887. [PMID: 38346599 DOI: 10.1016/j.ijpharm.2024.123887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 02/22/2024]
Abstract
Proteins acting as carriers in amorphous solid dispersions (ASDs) demonstrate a notable sensitivity to the spray drying process, potentially leading to changes in their conformation. The main aim of this study was to investigate the dissolution performance of ASDs based on proteins with different content of secondary structures, specifically β-sheet and α-helix structures. We prepared β-sheet-rich and α-helix-rich β-lactoglobulin (BLG), along with corresponding ASDs containing 10 wt% and 30 wt% drug loadings, through spray drying using celecoxib as the model drug. Circular dichroism and Fourier Transform Infrared Spectroscopy results revealed that even though changes in secondary structure were obtained in the spray-dried powders, the BLGs exhibited reversibility upon re-dissolving in phosphate buffer with varying pH levels. Both β-sheet-rich BLG and α-helix-rich BLG exhibited enhanced dissolution rates and higher solubility in the media with pH values far from the isoelectric point (pI) of BLG (pH 2, 7, 8, and 9) compared to the pH closer to the pI (pH 3, 4, 5, and 6). Notably, the release rate and solubility of the drug and BLG from both types of BLG-based ASDs at 10 wt% drug loading were largely dependent on the solubility of pure SD-BLGs. α-helix-rich BLG-ASDs consistently exhibited equivalent or superior performance to β-sheet-rich BLG-ASDs in terms of drug release rate and solubility, regardless of drug loading. Moreover, both types of BLG-based ASDs at 10 wt% drug loading exhibited faster release rates and higher solubility, for both the drug and BLG, compared to the ASDs at 30 wt% drug loading in pHs 2, 7, and 9 media.
Collapse
Affiliation(s)
- Xuezhi Zhuo
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Martina Tozzetti
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Anis Arnous
- Zerion Pharma A/S, Blokken 11, DK-3460 Birkerød, Denmark
| | - Donglei Leng
- Zerion Pharma A/S, Blokken 11, DK-3460 Birkerød, Denmark
| | - Vito Foderà
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Korbinian Löbmann
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark; Zerion Pharma A/S, Blokken 11, DK-3460 Birkerød, Denmark
| |
Collapse
|
43
|
Samsoen S, Dudognon É, Le Fer G, Fournier D, Woisel P, Affouard F. Impact of the polymer dispersity on the properties of curcumin/polyvinylpyrrolidone amorphous solid dispersions. Int J Pharm 2024; 653:123895. [PMID: 38346598 DOI: 10.1016/j.ijpharm.2024.123895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/20/2024] [Accepted: 02/07/2024] [Indexed: 02/19/2024]
Abstract
Amorphous solid dispersions (ASD) are known to enhance the absorption of poorly water-soluble drugs. In this work we synthesise well-defined Polyvinylpyrrolidone (PVP) to establish the impact of dispersity and chain-end functionality on the physical properties of Curcumin (CUR)/PVP ASD. Thermodynamic characterisation of synthesised PVP emphasises a strong effect of the dispersity on the glass transition temperature (Tg), 50 °C higher for synthesised PVP than for commercial PVP K12 of same molar mass. This increase of Tg affects the thermodynamic properties of CUR/PVP ASD successfully formulated up to 70 wt% of CUR by milling or solvent evaporation. The evolution of both the Tg and CUR solubility values versus CUR content points out the development of fairly strong CUR-PVP interactions that strengthen the antiplasticising effect of PVP on the Tg of ASD. However, for ASD formulated with commercial PVP this effect is counterbalanced at low CUR content by a plasticising effect due to the shortest PVP chains. Moreover, the overlay of the phase and state diagrams highlights the strong impact of the polymer dispersity on the stability of CUR/PVP ASD. ASD formulated with low dispersity PVP are stable on larger temperature and concentration ranges than those formulated with PVP K12.
Collapse
Affiliation(s)
- Simon Samsoen
- Univ. Lille, CNRS, INRAE, Centrale Lille, UMR 8207 - UMET - Unité Matériaux et Transformations, F-59000, Lille, France
| | - Émeline Dudognon
- Univ. Lille, CNRS, INRAE, Centrale Lille, UMR 8207 - UMET - Unité Matériaux et Transformations, F-59000, Lille, France.
| | - Gaëlle Le Fer
- Univ. Lille, CNRS, INRAE, Centrale Lille, UMR 8207 - UMET - Unité Matériaux et Transformations, F-59000, Lille, France.
| | - David Fournier
- Univ. Lille, CNRS, INRAE, Centrale Lille, UMR 8207 - UMET - Unité Matériaux et Transformations, F-59000, Lille, France
| | - Patrice Woisel
- Univ. Lille, CNRS, INRAE, Centrale Lille, UMR 8207 - UMET - Unité Matériaux et Transformations, F-59000, Lille, France
| | - Frédéric Affouard
- Univ. Lille, CNRS, INRAE, Centrale Lille, UMR 8207 - UMET - Unité Matériaux et Transformations, F-59000, Lille, France
| |
Collapse
|
44
|
Liu YS, Della Rocca J, Schenck L, Koynov A, Sifri RJ, Winston MS, Frank DS. Poly(vinylpyridine- co-vinylpyridine N-oxide) Excipients Mediate Rapid Dissolution and Sustained Supersaturation of Posaconazole Amorphous Solid Dispersions. Mol Pharm 2024; 21:1182-1191. [PMID: 38323546 DOI: 10.1021/acs.molpharmaceut.3c00789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
The chemical structure of excipients molecularly mixed in an amorphous solid dispersion (ASD) has a significant impact on properties of the ASD including dissolution behavior, physical stability, and bioavailability. Polymers used in ASDs require a balance between hydrophobic and hydrophilic functionalities to ensure rapid dissolution of the amorphous dispersion as well as sustained supersaturation of the drug in solution. This work demonstrates the use of postpolymerization functionalization of poly(vinylpyridine) excipients to elucidate the impact of polymer properties on the dissolution behavior of amorphous dispersions containing posaconazole. It was found that N-oxidation of pyridine functionalities increased the solubility of poly(vinylpyridine) derivatives in neutral aqueous conditions and allowed for nanoparticle formation which supplied posaconazole into solution at concentrations exceeding those achieved by more conventional excipients such as hydroxypropyl methylcellulose acetate succinate (HPMCAS) or Eudragit E PO. By leveraging these functional modifications of the parent poly(vinylpyridine) excipient to increase polymer hydrophilicity and minimize the effect of polymer on pH, a new polymeric excipient was optimized for rapid dissolution and supersaturation maintenance for a model compound.
Collapse
Affiliation(s)
- Yu-Sheng Liu
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Joseph Della Rocca
- Oral Formulation Sciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Luke Schenck
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Athanas Koynov
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Renee J Sifri
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Matthew S Winston
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Derek S Frank
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| |
Collapse
|
45
|
Queiroz LHS, Barros RS, de Sousa FF, Lage MR, Sarraguça MC, Ribeiro PRS. Preparation and Characterization of a Rifampicin Coamorphous Material with Tromethamine Coformer: An Experimental-Theoretical Study. Mol Pharm 2024; 21:1272-1284. [PMID: 38361428 DOI: 10.1021/acs.molpharmaceut.3c00947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Rifampicin (RIF) is an antibiotic used to treat tuberculosis and leprosy. Even though RIF is a market-available drug, it has a low aqueous solubility, hindering its bioavailability. Among the strategies for bioavailability improvement of poorly soluble drugs, coamorphous systems have been revealed as an alternative in the increase of the aqueous solubility of drug systems and at the same time also increasing the amorphous state stability and dissolution rate when compared with the neat drug. In this work, a new coamorphous form from RIF and tromethamine (TRIS) was synthesized by slow evaporation. Structural, electronic, and thermodynamic properties and solvation effects, as well as drug-coformer intermolecular interactions, were studied through density functional theory (DFT) calculations. Powder X-ray diffraction (PXRD) data allowed us to verify the formation of a new coamorphous. In addition, the DFT study indicates a possible intermolecular interaction by hydrogen bonds between the available amino and carbonyl groups of RIF and the hydroxyl and amino groups of TRIS. The theoretical spectra obtained are in good agreement with the experimental data, suggesting the main interactions occurring in the formation of the coamorphous system. PXRD was used to study the physical stability of the coamorphous system under accelerated ICH conditions (40 °C and 75% RH), indicating that the material remained in an amorphous state up to 180 days. The thermogravimetry result of this material showed a good thermal stability up to 153 °C, and differential scanning calorimetry showed that the glass temperature (Tg) was at 70.0 °C. Solubility studies demonstrated an increase in the solubility of RIF by 5.5-fold when compared with its crystalline counterpart. Therefore, this new material presents critical parameters that can be considered in the development of new coamorphous formulations.
Collapse
Affiliation(s)
- Luís H S Queiroz
- NUPFARQ, Programa de Pós-Graduação em Ciência dos Materiais (PPGCM), Centro de Ciências de Imperatriz (CCIM), Universidade Federal do Maranhão (UFMA), Imperatriz, Maranhão 65.900-410, Brazil
| | - Ranna S Barros
- NUPFARQ, Programa de Pós-Graduação em Ciência dos Materiais (PPGCM), Centro de Ciências de Imperatriz (CCIM), Universidade Federal do Maranhão (UFMA), Imperatriz, Maranhão 65.900-410, Brazil
| | - Francisco F de Sousa
- Instituto de Ciências Exatas e Naturais, Universidade Federal do Pará (UFPA), Belém, Pará 65.075-110, Brazil
| | - Mateus R Lage
- NUPFARQ, Programa de Pós-Graduação em Ciência dos Materiais (PPGCM), Centro de Ciências de Imperatriz (CCIM), Universidade Federal do Maranhão (UFMA), Imperatriz, Maranhão 65.900-410, Brazil
- Coordenação do Curso de Ciência e Tecnologia, Centro de Ciências de Balsas, Universidade Federal do Maranhão (UFMA), Balsas, Maranhão 65.800-000, Brazil
| | - Mafalda C Sarraguça
- LAQV, REQUIMTE, Department of Chemical Sciences, Laboratory of Applied Chemistry, Faculty of Pharmacy, Porto University, Rua de Jorge Viterbo Ferreira, 228, Porto 4050-313, Portugal
| | - Paulo R S Ribeiro
- NUPFARQ, Programa de Pós-Graduação em Ciência dos Materiais (PPGCM), Centro de Ciências de Imperatriz (CCIM), Universidade Federal do Maranhão (UFMA), Imperatriz, Maranhão 65.900-410, Brazil
| |
Collapse
|
46
|
Zhang Z, Cui Z, Zhang J, Zheng H, Zhou Z, Wu Z, Wang Z, Fu B. Remineralizing effects of hydroxypropyl methylcellulose film-loaded amorphous calcium phosphate nanoprecursors on enamel artificial caries lesions. J Mech Behav Biomed Mater 2024; 151:106408. [PMID: 38244421 DOI: 10.1016/j.jmbbm.2024.106408] [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: 11/23/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 01/22/2024]
Abstract
OBJECTIVES This study was to investigate hydroxypropyl methylcellulose (HPMC) film as a carrier for amorphous fluorinated calcium phosphate (AFCP) nanoprecursors to continuously deliver biomimetic remineralization of enamel artificial caries lesions (ACL). MATERIALS AND METHODS The AFCP/HPMC films were comprised of 25 wt% AFCP nanoparticles and 75 wt% HPMC. They were characterized by transmission electron microscopy (TEM), Fourier transform infrared spectroscopy (FTIR), X-ray diffraction (XRD), X-ray photoelectron spectroscopy (XPS), and biocompatibility tests. Forty enamel ACL were prepared and randomly divided into four groups (n = 10): The enamel surfaces were covered with a pure HPMC film, Tooth Mousse Plus (contains 10% CPP-ACP and 0.2% NaF), and AFCP/HPMC film, or without any things (serving as negative control). Subsequently, all samples were alternatively kept in artificial saliva and a modified pH-cycling before they were characterized by Micro-CT, scanning electron microscopy (SEM), energy-dispersive X-ray spectroscopy (EDX), attenuated total reflectance (ATR)-FTIR, XRD, and nanoindentation. RESULTS After the enamel ACL was challenged by pH cycling, Tooth Mousse Plus and AFCP/HPMC film groups exhibited less lesion depth and mineral loss than the negative control and pure HPMC film groups. Additionally, the AFCP/HPMC film group revealed a highest remineralization rate of 55.34 ± 3.10 % among the all groups (p < 0.001). The SEM findings showed that the enamel ACL were densely deposited with minerals in the AFCP/HPMC film group, and the EDX results suggested a higher content of fluorine in the remineralized tissues. In particular, the AFCP/HPMC film group exhibited the best nanomechanical performance after 2 weeks of pH cycling (p < 0.05), with the hardness (H) restored from 0.29 ± 0.19 to 2.69 ± 0.70 GPa, and elastic modulus (Er) restored from 10.77 ± 5.30 to 68.83 ± 12.72 GPa. CONCLUSION The AFCP/HPMC film might be used as a promising strategy for arresting or reversing incipient enamel caries lesions.
Collapse
Affiliation(s)
- Zhixin Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China; Department of Stomatology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, 362000, Fujian, China.
| | - Zihan Cui
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Jianping Zhang
- Department of Orthopaedics, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, 363000, Fujian, China
| | - Haiyan Zheng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Zihuai Zhou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Zhifang Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Zhe Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China.
| | - Baiping Fu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China.
| |
Collapse
|
47
|
Hofmann N, Harms M, Mäder K. ASDs of PROTACs: Spray-dried solid dispersions as enabling formulations. Int J Pharm 2024; 650:123725. [PMID: 38113976 DOI: 10.1016/j.ijpharm.2023.123725] [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: 09/25/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 12/21/2023]
Abstract
Proteolysis targeting chimeras (PROTACs) are a promising class of pharmaceutical agents with a unique mode of action. PROTACs enable the targeting of a broad variety of structures including transcription factors and other "undruggable" targets. The poor solubility and slow dissolution of PROTACs currently limit the extensive use of their potential. Up to date, only very limited drug delivery options have been examined to address this challenge. Therefore, we explored the potential of amorphous solid dispersions (ASDs) by spray drying a model PROTAC with different polymers. The resulting formulations were assessed in terms of purity, solid state, dissolution performance, and stability. A strong increase in supersaturation compared to the physical mixture was provided, although in both systems the PROTAC molecule itself was already in the amorphous state. Evaluation of the reasons for the superiority of the ASD formulations revealed that the major factor was the homogeneous, molecular distribution of the active pharmaceutical ingredient (API) in the polymer matrix, as well as improved wettability of the formulation containing Soluplus compared to the physical mixture. The manufactured formulations were stable over a minimum of 8 weeks when protected from light and humidity.
Collapse
Affiliation(s)
- Nicole Hofmann
- Global Drug Product Development, Orals Development, Merck KGaA, Frankfurter Straße 250, 64293 Darmstadt, Germany; Institute of Pharmacy, Faculty I of Natural Sciences, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Strasse 3, 06120 Halle (Saale), Germany
| | - Meike Harms
- Global Drug Product Development, Orals Development, Merck KGaA, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Karsten Mäder
- Institute of Pharmacy, Faculty I of Natural Sciences, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Strasse 3, 06120 Halle (Saale), Germany.
| |
Collapse
|
48
|
Kayalar C, Helal N, Mohamed EM, Dharani S, Khuroo T, Kuttolamadom MA, Rahman Z, Khan MA. In Vitro and In Vivo testing of 3D-Printed Amorphous Lopinavir Printlets by Selective Laser Sinitering: Improved Bioavailability of a Poorly Soluble Drug. AAPS PharmSciTech 2024; 25:20. [PMID: 38267637 DOI: 10.1208/s12249-023-02729-y] [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: 08/29/2023] [Accepted: 12/19/2023] [Indexed: 01/26/2024] Open
Abstract
The aim of this paper was to investigate the effects of formulation parameters on the physicochemical and pharmacokinetic (PK) behavior of amorphous printlets of lopinavir (LPV) manufactured by selective laser sintering 3D printing method (SLS). The formulation variables investigated were disintegrants (magnesium aluminum silicate at 5-10%, microcrystalline cellulose at 10-20%) and the polymer (Kollicoat® IR at 42-57%), while keeping printing parameters constant. Differential scanning calorimetry, X-ray powder diffraction, and Fourier-transform infrared analysis confirmed the transformation of the crystalline drug into an amorphous form. A direct correlation was found between the disintegrant concentration and dissolution. The dissolved drug ranged from 71.1 ± 5.7% to 99.3 ± 2.7% within 120 min. A comparative PK study in rabbits showed significant differences in the rate and extent of absorption between printlets and compressed tablets. The values for Tmax, Cmax, and AUC were 4 times faster, and 2.5 and 1.7 times higher in the printlets compared to the compressed tablets, respectively. In conclusion, the SLS printing method can be used to create an amorphous delivery system through a single continuous process.
Collapse
Affiliation(s)
- Canberk Kayalar
- Irma Lerma Rangel School of Pharmacy, Texas A&M Health Science Center, Texas A&M University, Reynolds Medical Sciences Building, Suite 159, College Station, TX, 77843-1114, United States of America
| | - Nada Helal
- Irma Lerma Rangel School of Pharmacy, Texas A&M Health Science Center, Texas A&M University, Reynolds Medical Sciences Building, Suite 159, College Station, TX, 77843-1114, United States of America
| | - Eman M Mohamed
- Irma Lerma Rangel School of Pharmacy, Texas A&M Health Science Center, Texas A&M University, Reynolds Medical Sciences Building, Suite 159, College Station, TX, 77843-1114, United States of America
| | - Sathish Dharani
- Irma Lerma Rangel School of Pharmacy, Texas A&M Health Science Center, Texas A&M University, Reynolds Medical Sciences Building, Suite 159, College Station, TX, 77843-1114, United States of America
| | - Tahir Khuroo
- Irma Lerma Rangel School of Pharmacy, Texas A&M Health Science Center, Texas A&M University, Reynolds Medical Sciences Building, Suite 159, College Station, TX, 77843-1114, United States of America
| | - Mathew A Kuttolamadom
- Dept. of Engineering Technology & Industrial Distribution, College of Engineering, Texas A&M University, College Station, TX, 77843, United States of America
| | - Ziyaur Rahman
- Irma Lerma Rangel School of Pharmacy, Texas A&M Health Science Center, Texas A&M University, Reynolds Medical Sciences Building, Suite 159, College Station, TX, 77843-1114, United States of America
| | - Mansoor A Khan
- Irma Lerma Rangel School of Pharmacy, Texas A&M Health Science Center, Texas A&M University, Reynolds Medical Sciences Building, Suite 159, College Station, TX, 77843-1114, United States of America.
| |
Collapse
|
49
|
Aulifa DL, Al Shofwan AA, Megantara S, Fakih TM, Budiman A. Elucidation of Molecular Interactions Between Drug-Polymer in Amorphous Solid Dispersion by a Computational Approach Using Molecular Dynamics Simulations. Adv Appl Bioinform Chem 2024; 17:1-19. [PMID: 38282640 PMCID: PMC10821732 DOI: 10.2147/aabc.s441628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 01/16/2024] [Indexed: 01/30/2024] Open
Abstract
Introduction Amorphous drug dispersion is frequently used to enhance the solubility and dissolution of poorly water-soluble drugs, thereby improving their oral bioavailability. The dispersion of these drugs into polymer matrix can inhibit their recrystallization. The inter-molecular interactions between drug and polymer plays a role in the improvement of the dissolution rate, solubility, and physical stability of drug. Aim This study aims to investigate the formation and interactions of ritonavir (RTV)/poloxamer (PLX) amorphous formulation using a computational approach via molecular dynamics (MD) simulations, which mimicked solvent evaporation and melt-quenching method. Methods TheRoot Mean Square Deviation (RMSD) value, Root Mean Square Fluctuation (RMSF), Radial Distribution Function (RDF), Radius of Gyration (Rg), Solvent Accessible Surface Area (SASA), and hydrogen bond interactions were analyzed to determine interaction mechanisms between RTV and PLX in amorphous solid dispersion. Results The pi-alkyl bonds between RTV and PLX were formed after simulations of solvent evaporation, while the hydrogen bond interactions of RTV-PLX was observed during melt method simulations. These results indicate the successful formulation of amorphous solid dispersion (ASD) from RTV and PLX. The RMSD values obtained from the solvent evaporation, melt-cooling-A, melt-cooling-B, and melt-cooling-C methods were 3.33 Å, 1.97 Å, 1.30 Å, and 1.29 Å, respectively, while the average RMSF values were 2.65 Å, 1.04 Å, 1.05 Å, and 1.07 Å, respectively. This indicates that the suppression of translational motion of RTV from the melt method can be stronger than solvent evaporation caused by the intermolecular interactions of RTV-PLX. Conclusion MD simulations helped in understanding the formation and interaction mechanisms of ASD formulations that were difficult to detect by experimental approaches.
Collapse
Affiliation(s)
- Diah Lia Aulifa
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Adnan Aly Al Shofwan
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Sandra Megantara
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Taufik Muhammad Fakih
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, Universitas Islam Bandung, Bandung, Indonesia
| | - Arif Budiman
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| |
Collapse
|
50
|
Hasegawa K, Goto S, Kataoka H, Chatani H, Kinoshita T, Yokoyama H, Tsuchida T. Quantification of crystallinity during indomethacin crystalline transformation from α- to γ-polymorphic forms and of the thermodynamic contribution to dissolution in aqueous buffer and solutions of solubilizer. RSC Adv 2024; 14:4129-4141. [PMID: 38292264 PMCID: PMC10825737 DOI: 10.1039/d3ra08481g] [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: 12/12/2023] [Accepted: 01/19/2024] [Indexed: 02/01/2024] Open
Abstract
The thermodynamic properties and dissolution of indomethacin (INM) were analyzed as models for poorly water-soluble drugs. Physical mixtures of the most stable γ-form and metastable α-form of INM at various proportions were prepared, and their individual signal intensities proportional to their mole fractions were observed using X-ray powder diffraction and Fourier transform infrared spectrometry at standard temperature. The endothermic signals of the α-form, with a melting point of 426 K, and that of the γ-form, with a melting point of 433 K, were obtained by differential scanning calorimetry (DSC). Furthermore, an exothermic DSC peak of the α/γ-phase transition at approximately 428 K was obtained. As we computed the melting entropy of the α-form and that of its transformation, the frequency of the transition was quantitatively determined, which indicated the maximum of the α/γ-phase transition at an α-form proportion of 68%. Subsequently, the thermodynamic contributions of the α- and γ-forms were analyzed using a Van't Hoff plot for solubility in aqueous solutions at pH 6.8. The dissolution enthalpies for α- and γ-forms were 28.2 and 31.2 kJ mol-1, respectively, which are in agreement with the quantitative contribution predicted by the product of the temperature and melting entropy. The contribution of melting entropy was conserved in different dissolution processes with aqueous solvents containing lidocaine, diltiazem, l-carnosine, and aspartame as solubilizers; their γ-form Setschenow coefficients were -39.6, +82.9, -17.3, and +23.2, whereas those of the α-form were -39.7, +80.4, -16.7, and +22.7, respectively. We conclude that the dissolution ability of the solid state and solubilizers indicate their additivity independently.
Collapse
Affiliation(s)
- Kanji Hasegawa
- Faculty of Pharmaceutical Sciences, Tokyo University of Science 2641 Yamazaki, Noda Chiba 278-8510 Japan
| | - Satoru Goto
- Faculty of Pharmaceutical Sciences, Tokyo University of Science 2641 Yamazaki, Noda Chiba 278-8510 Japan
| | - Hikaru Kataoka
- Faculty of Pharmaceutical Sciences, Tokyo University of Science 2641 Yamazaki, Noda Chiba 278-8510 Japan
| | - Hitoshi Chatani
- Faculty of Pharmaceutical Sciences, Tokyo University of Science 2641 Yamazaki, Noda Chiba 278-8510 Japan
| | - Takatoshi Kinoshita
- Faculty of Pharmaceutical Sciences, Tokyo University of Science 2641 Yamazaki, Noda Chiba 278-8510 Japan
| | - Hideshi Yokoyama
- Faculty of Pharmaceutical Sciences, Tokyo University of Science 2641 Yamazaki, Noda Chiba 278-8510 Japan
| | - Tomohiro Tsuchida
- Faculty of Pharmaceutical Sciences, Tokyo University of Science 2641 Yamazaki, Noda Chiba 278-8510 Japan
| |
Collapse
|