Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.231
Filtrer
1.
Mol Pharm ; 21(7): 3395-3406, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38836777

RÉSUMÉ

The incorporation of a counterion into an amorphous solid dispersion (ASD) has been proven to be an attractive strategy to improve the drug dissolution rate. In this work, the generality of enhancing the dissolution rates of free acid ASDs by incorporating sodium hydroxide (NaOH) was studied by surface-area-normalized dissolution. A set of diverse drug molecules, two common polymer carriers (copovidone or PVPVA and hydroxypropyl methylcellulose acetate succinate or HPMCAS), and two sample preparation methods (rotary evaporation and spray drying) were investigated. When PVPVA was used as the polymer carrier for the drugs in this study, enhancements of dissolution rates from 7 to 78 times were observed by the incorporation of NaOH into the ASDs at a 1:1 molar ratio with respect to the drug. The drugs having lower amorphous solubilities showed greater enhancement ratios, providing a promising path to improve the drug release performance from their ASDs. Samples generated by rotary evaporation and spray drying demonstrated comparable dissolution rates and enhancements when NaOH was added, establishing a theoretical foundation to bridge the ASD dissolution performance for samples prepared by different solvent-removal processes. In the comparison of polymer carriers, when HPMCAS was applied in the selected system (indomethacin ASD), a dissolution rate enhancement of 2.7 times by the incorporated NaOH was observed, significantly lower than the enhancement of 53 times from the PVPVA-based ASD. This was attributed to the combination of a lower dissolution rate of HPMCAS and the competition for NaOH between IMC and HPMCAS. By studying the generality of enhancing ASD dissolution rates by the incorporation of counterions, this study provides valuable insights into further improving drug release from ASD formulations of poorly water-soluble drugs.


Sujet(s)
Libération de médicament , Méthylcellulose , Hydroxyde de sodium , Solubilité , Hydroxyde de sodium/composition chimique , Méthylcellulose/composition chimique , Méthylcellulose/analogues et dérivés , Polymères/composition chimique , Vecteurs de médicaments/composition chimique , Chimie pharmaceutique/méthodes , Préparation de médicament/méthodes , Pyrrolidines/composition chimique
2.
Mol Pharm ; 21(7): 3375-3382, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38885189

RÉSUMÉ

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.


Sujet(s)
Polymères , Polymères/composition chimique , Méthylcellulose/composition chimique , Méthylcellulose/analogues et dérivés , Cristallisation/méthodes , Cellulose/composition chimique , Cellulose/analogues et dérivés , Résines acryliques/composition chimique , Sels/composition chimique , Dérivés de l'hypromellose/composition chimique , Solubilité
3.
ACS Biomater Sci Eng ; 10(6): 3833-3841, 2024 06 10.
Article de Anglais | MEDLINE | ID: mdl-38747490

RÉSUMÉ

This study explores the realm of personalized medicine by investigating the utilization of 3D-printed dosage forms, specifically focusing on patient-specific enteric capsules designed for the modified release of ketoprofen, serving as a model drug. The research investigates two distinct scenarios: the modification of drug release from 3D-printed capsules crafted from hydroxypropyl methylcellulose phthalate:polyethylene glycol (HPMCP:PEG) and poly(vinyl alcohol) (PVA), tailored for pH sensitivity and delayed release modes, respectively. Additionally, a novel ketoprofen-loaded self-nanoemulsifying drug delivery system (SNEDDS) based on pomegranate seed oil (PSO) was developed, characterized, and employed as a fill material for the capsules. Through the preparation and characterization of the HPMCP:PEG based filament via the hot-melt extrusion method, the study thoroughly investigated its thermal and mechanical properties. Notably, the in vitro drug release analysis unveiled the intricate interplay between ketoprofen release, polymer type, and capsule thickness. Furthermore, the incorporation of ketoprofen into the SNEDDS exhibited an enhancement in its in vitro cylooxygenase-2 (COX-2) inhibitory activity. These findings collectively underscore the potential of 3D printing in shaping tailored drug delivery systems, thereby contributing significantly to the advancement of personalized medicine.


Sujet(s)
Capsules , Libération de médicament , Émulsions , Kétoprofène , Médecine de précision , Impression tridimensionnelle , Kétoprofène/composition chimique , Médecine de précision/méthodes , Humains , Émulsions/composition chimique , Polyéthylène glycols/composition chimique , Systèmes de délivrance de médicaments/méthodes , Préparations à action retardée , Méthylcellulose/composition chimique , Méthylcellulose/analogues et dérivés , Poly(alcool vinylique)/composition chimique
4.
Mol Pharm ; 21(6): 2908-2921, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38743928

RÉSUMÉ

The physical stability of amorphous solid dispersions (ASDs) is a major topic in the formulation research of oral dosage forms. To minimize the effort of investigating the long-term stability using cost- and time-consuming experiments, we developed a thermodynamic and kinetic modeling framework to predict and understand the crystallization kinetics of ASDs during long-term storage below the glass transition. Since crystallization of the active phrarmaceutical ingredients (APIs) in ASDs largely depends on the amount of water absorbed by the ASDs, water-sorption kinetics and API-crystallization kinetics were considered simultaneously. The developed modeling approach allows prediction of the time evolution of viscosity, supersaturation, and crystallinity as a function of drug load, relative humidity, and temperature. It was applied and evaluated against two-year-lasting crystallization experiments of ASDs containing nifedipine and copovidone or HPMCAS measured in part I of this work. We could show that the proposed modeling approach is able to describe the interplay between water sorption and API crystallization and to predict long-term stabilities of ASDs just based on short-term measurements. Most importantly, it enables explaining and understanding the reasons for different and sometimes even unexpected crystallization behaviors of ASDs.


Sujet(s)
Cristallisation , Eau , Cristallisation/méthodes , Eau/composition chimique , Cinétique , Stabilité de médicament , Nifédipine/composition chimique , Composés vinyliques/composition chimique , Thermodynamique , Pyrrolidines/composition chimique , Viscosité , Chimie pharmaceutique/méthodes , Humidité , Température , Solubilité , Méthylcellulose/composition chimique , Méthylcellulose/analogues et dérivés
5.
J Colloid Interface Sci ; 669: 975-983, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38759596

RÉSUMÉ

HYPOTHESIS: Hydroxypropyl methylcellulose phthalate (HPMCP) is an enteric polymer that has been employed in drug delivery systems to delay the release of the encapsulated active pharmaceutical ingredients through its pH-responsive solubility change. This has been recently demonstrated as an effective means for delaying the drug release from gelatin/HPMCP hydrogels at gastric pH values. However, structural characteristics of HPMCP agglomeration in gelatin/HPMCP hydrogels is not well understood thus limiting further tailoring of their material properties. EXPERIMENTS: We investigated the multiscale structure of a gelatin/HPMCP hydrogel (1:1 by weight) between pH 2 and 6 at 37 °C, i.e. above the upper critical solution transition temperature of gelatin, using small-angle X-ray scattering and contrast-variation small-angle neutron scattering to understand the pH-responsive structure of HPMCP and the cross-correlation between gelatin and HPMCP. FINDINGS: Agglomeration of HPMCP between pH 2 and 4 was evidenced by the formation of mass fractal structures, with a fractal dimension ranging from 1.5 to 2.7, comprising primary particles with a radius of gyration ranging from 70 to 140 Å. Blending with gelatin influenced the fractal structure of HPMCP and the primary particle size. Gelatin and HPMCP exhibited negative cross-correlation in all probed length scales and pH values, which was attributed to volume-exclusion interaction in a double-network-like solution architecture.


Sujet(s)
Gélatine , Méthylcellulose , Taille de particule , Diffusion aux petits angles , Gélatine/composition chimique , Concentration en ions d'hydrogène , Méthylcellulose/composition chimique , Méthylcellulose/analogues et dérivés , Hydrogels/composition chimique , Structure moléculaire
6.
Mol Pharm ; 21(3): 1466-1478, 2024 Mar 04.
Article de Anglais | MEDLINE | ID: mdl-38346390

RÉSUMÉ

The interplay between drug and polymer chemistry and its impact on drug release from an amorphous solid dispersion (ASD) is a relatively underexplored area. Herein, the release rates of several drugs of diverse chemistry from hydroxypropyl methylcellulose acetate succinate (HPMCAS)-based ASDs were explored using surface area normalized dissolution. The tendency of the drug to form an insoluble complex with HPMCAS was determined through coprecipitation experiments. The role of pH and the extent of drug ionization were probed to evaluate the role of electrostatic interactions in complex formation. Relationships between the extent of complexation and the drug release rate from an ASD were observed, whereby the drugs could be divided into two groups. Drugs with a low extent of insoluble complex formation with HPMCAS tended to be neutral or anionic and showed reasonable release at pH 6.8 even at higher drug loadings. Cationic drugs formed insoluble complexes with HPMCAS and showed poor release when formulated as an ASD. Thus, and somewhat counterintuitively, a weakly basic drug showed a reduced release rate from an ASD at a bulk solution pH where it was ionized, relative to when unionized. The opposite trend was observed in the absence of polymer for the neat amorphous drug. In conclusion, electrostatic interactions between HPMCAS and lipophilic cationic drugs led to insoluble complex formation, which in turn resulted in ASDs with poor release performance.


Sujet(s)
Méthylcellulose , Méthylcellulose/analogues et dérivés , Polymères , Polymères/composition chimique , Solubilité , Libération de médicament , Méthylcellulose/composition chimique
7.
Int J Biol Macromol ; 261(Pt 1): 129701, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38280709

RÉSUMÉ

The development of flexible and porous materials to control antibacterial delivery is a pivotal endeavor in medical science. In this study, we aimed to produce long and defect-free fibers made of zein and hydroxypropyl methylcellulose acetate succinate (HPMCAS) to be used as a platform for the release of metronidazole (MDZ) and metronidazole benzoate (BMDZ) to be potentially used in periodontal treatment. Microfibers prepared via electrospinning under a 2:3 (w/w) zein to HPMCAS ratio, containing 0.5 % (w/w) poly(ethylene oxide) (PEO) and 1 % (w/w) cellulose nanofibril (CNF) were loaded with 40 % (w/w) MDZ, 40 % (w/w) BMDZ, or a combination of 20 % (w/w) of each drug. The addition of CNF improved the electrospinning process, resulting in long fibers with reduced MDZ and BMDZ surface crystallization. MDZ- and BMDZ-incorporated fibers were semicrystalline and displayed commendable compatibility among drugs, nanocellulose and polymeric chains. Release tests showed that zein/HPMCAS/PEO fibers without CNF and with 20 % (w/w) MDZ/ 20 % (w/w) BMDZ released the drug at a slower and more sustained rate compared to other samples over extended periods (up to 5 days), which is a favorable aspect concerning periodontitis treatment.


Sujet(s)
Méthylcellulose/analogues et dérivés , Métronidazole , Zéine , Métronidazole/pharmacologie , Cellulose , Benzoates
8.
Eur J Pharm Biopharm ; 177: 289-307, 2022 Aug.
Article de Anglais | MEDLINE | ID: mdl-35872180

RÉSUMÉ

Amorphous solid dispersions (ASDs) are a proven system for achieving a supersaturated state of drug, in which the concentration of drug is greater than its crystalline solubility. The usage of Hydroxypropyl Methylcellulose Acetate Succinate (HPMCAS) in the development of ASDs has grown significantly, as evidenced by the fact that majority of commercially approved ASD formulations are based on HPMCAS. HPMCAS has been widely utilized as a solubility enhancer and precipitation inhibitor or stabilizer to achieve supersaturation and inhibit crystallization of drugs in the gastrointestinal tract. The characteristics of HPMCAS ASDs such as less hygroscopic, strong drug-polymer hydrophobic interactions, high solubilization efficiency, greater potential to generate, maintain drug supersaturation and crystallization inhibition outperform other polymeric carriers in ASD development. Furthermore, combining HPMCAS with other polymers or surfactants as ternary ASDs could be a viable approach for enhancing oral absorption of poorly soluble drugs. This review discusses the concepts of supersaturation maintenance or precipitation inhibition of HPMCAS in the ASD formulations. In addition, the mechanisms underlying for improved dissolution performance, oral bioavailability and stability of HPMCAS ASDs are explored.


Sujet(s)
Méthylcellulose , Polymères , Préparation de médicament , Méthylcellulose/analogues et dérivés , Méthylcellulose/composition chimique , Polymères/composition chimique , Solubilité
9.
Pharm Dev Technol ; 27(5): 615-624, 2022 Jun.
Article de Anglais | MEDLINE | ID: mdl-35786299

RÉSUMÉ

Zidovudine (AZT) has been widely used alone or in combination with other antiretroviral drugs for the treatment of human immunodeficiency virus. Its erratic oral bioavailability necessitates frequent administration of high doses, resulting in severe side effects. In this study, the design of mucoadhesive solid dispersions (SDs) based on chitosan (CS) and hypromellose phthalate (HP) was rationalized as a potential approach to modulate AZT physicochemical and pharmaceutical properties. SDs were prepared at different drug:polymer ratios, using an eco-friendly technique, which avoids the use of organic solvents. Particles with diameter from 56 to 73 µm and negative zeta potentials (-27 to -32 mV) were successfully prepared, achieving high drug content. Infrared spectroscopy revealed interactions between polymers but no interactions between the polymers and AZT. Calorimetry and X-ray diffraction analyses showed that AZT was amorphized into the SDs. The mucoadhesive properties of SDs were evidenced, and the control of AZT release rates from the matrix was achieved, mainly in acid media. The simple, low-cost, and scalable technology proposed for production of SDs as a carrier platform for AZT is an innovative approach, and it proved to be a feasible strategy for modulation the physico-chemical, mucoadhesive, and release properties of the drug.


Sujet(s)
Chitosane , Chitosane/composition chimique , Vecteurs de médicaments/composition chimique , Humains , Dérivés de l'hypromellose , Méthylcellulose/analogues et dérivés , Polymères/composition chimique , Solubilité , Zidovudine/composition chimique
10.
Assay Drug Dev Technol ; 20(4): 149-163, 2022.
Article de Anglais | MEDLINE | ID: mdl-35723615

RÉSUMÉ

Poorly aqueous soluble active pharmaceutical ingredients are highly risky development candidates and remain a concern of pharmaceutical industries in drug discovery and development processes. Pharmaceutical industries are putting significant efforts into the target identification and lead candidate development using combinatorial chemistry. About 40% of compounds arising from combinatorial screening are poorly water soluble. Pharmaceutical industries evolved over this challenge by coming up with reproducible and scalable particle size reduction or by identifying alternate morphs. Another important area where pharmaceutical industries are working is solid dispersion technology. With the emergence of the hot-melt extrusion and spray drying approach, many molecules have been brought to the market using solid dispersion technology from the discovery phase by improving bioavailability and thereby efficacy. Although the solid solution technology in the last 60 years evolved from eutectic mixtures, solid dispersions using water-soluble polymers, and enteric polymers especially hydroxypropyl methylcellulose acetate succinate (HPMCAS), still there is no preformulation tool to identify correct polymer or polymer combination at the early stage of development. Thus, this leads to the urgent need to focus on the design and development of third-generation solid dispersions for the unmet needs of the industries and society.


Sujet(s)
Vecteurs de médicaments , Polymères , Vecteurs de médicaments/composition chimique , Préparation de médicament , Méthylcellulose/analogues et dérivés , Polymères/composition chimique , Solubilité , Eau
11.
ACS Infect Dis ; 8(3): 472-481, 2022 03 11.
Article de Anglais | MEDLINE | ID: mdl-35230825

RÉSUMÉ

Salmonella enterica subsp. enterica serovar Enteritidis (S. Enteritidis) in poultry is most often transmitted by the fecal-oral route, which can be attributed to high population density. Upon encountering the innate immune response in a host, the pathogen triggers a stress response and virulence factors to help it survive in the host. The aim of this study was to evaluate the effect of hypromellose acetate/succinate (HPMCAS)-coated alginate microparticles containing the Ctx(Ile21)-Ha antimicrobial peptide (AMP) on both intestinal colonization and systemic infection of laying hens challenged with S. Enteritidis. The applied AMP microsystem reduced the bacterial load of S. Enteritidis in the liver, with a statistical significance between groups A (control, no Ctx(Ile21)-Ha peptide) and B (2.5 mg of Ctx(Ile21)-Ha/kg) at 2 days postinfection (dpi), potentially indicating the effectiveness of Ctx(Ile21)-Ha in the first stage of infection by S. Enteritidis. In addition, the results showed a significant decrease in the S. Enteritidis counts in the spleen and cecal content at 5 dpi; remarkably, no S. Enteritidis counts were observed in livers at 5, 7, and 14 dpi, regardless of the Ctx(Ile21)-Ha dosage (p-value <0.0001). Using the Chi-square test, the effect of AMP microparticles on S. Enteritidis fecal excretion was also evaluated, and a significantly lower bacterial excretion was observed over 21 days in groups B and C, in comparison with the untreated control (p-value <0.05). In summary, the use of HPMCAS-Ctx(Ile21)-Ha peptide microcapsules in laying hens drastically reduced the systemic infection of S. Enteritidis, mainly in the liver, indicating a potential for application as a feed additive against this pathogen.


Sujet(s)
Anti-infectieux , Salmonelloses animales , Alginates , Animaux , Poulets/microbiologie , Poulets/physiologie , Femelle , Méthylcellulose/analogues et dérivés , Salmonelloses animales/traitement médicamenteux , Salmonelloses animales/microbiologie , Salmonella enteritidis/physiologie
12.
Int J Pharm ; 616: 121553, 2022 Mar 25.
Article de Anglais | MEDLINE | ID: mdl-35131354

RÉSUMÉ

3D printing (3DP) by fused deposition modelling (FDM) is one of the most extensively developed methods in additive manufacturing. Optimizing printability by improving feedability, nozzle extrusion, and layer deposition is crucial for manufacturing solid oral dosage forms with desirable properties. This work aimed to use HPMCAS (AffinisolTM HPMCAS 716) to prepare filaments for FDM-3DP using hot-melt extrusion (HME). It explored and demonstrated the effect of HME-filament composition and fabrication on printability by evaluating thermal, mechanical, and thermo-rheological properties. It also showed that the HME-Polymer filament composition used in FDM-3DP manufacture of oral solid dosage forms provides a tailored drug release profile. HME (HAAKE MiniLab) and FDM-3DP (MakerBot) were used to prepare HME-filaments and printed objects, respectively. Two diverse ways of improving the mechanical properties of HME-filaments were deduced by changing the formulation to enable feeding through the roller gears of the printer nozzle. These include plasticizing the polymer and adding an insoluble structuring agent (talc) into the formulation. Experimental feedability was predicted using texture analysis results was a function of PEG concentration, and glass-transition temperature (Tg) values of HME-filaments. The effect of high HME screw speed (100 rpm) resulted in inhomogeneity of HME-filament, which resulted in inconsistency of the printer nozzle extrudate and printed layers. The variability of the glass-transition temperature (Tg) of the HME-filament supported by scanning electron microscopy (SEM) images of nozzle extrudates and the lateral wall of the printed tablet helped explain this result. The melt viscosity of HPMCAS formulations was investigated using a capillary rheometer. The high viscosity of unplasticized HPMCAS was concluded to be an additional restriction for nozzle extrusion. The plasticization of HPMCAS and the addition of talc into the formulation were shown to improve thickness consistency of printed layers (using homogeneous HME-filaments). A good correlation (R2 = 0.9546) between the solidification threshold (low-frequency oscillation test determined by parallel-plate rheometer) and Tg of HME-filaments was also established. Drug-loaded and placebo HPMCAS-based formulations were shown to be successfully printed, with the former providing tailored drug release profiles based on variation of internal geometry (infill).


Sujet(s)
Excipients , Technologie pharmaceutique , Formes posologiques , Libération de médicament , Méthylcellulose/analogues et dérivés , Impression tridimensionnelle , Comprimés , Technologie pharmaceutique/méthodes
13.
Int J Pharm ; 615: 121471, 2022 Mar 05.
Article de Anglais | MEDLINE | ID: mdl-35041915

RÉSUMÉ

Nucleation inhibition and maintenance of drug supersaturation over a prolonged period are desirable for improving oral absorption of amorphous solid dispersions. The present study investigates the impact of binary and ternary amorphous solid dispersions on the supersaturation kinetics of nifedipine using the polymers hydroxypropylmethylcellulose acetate succinate (HPMCAS) LG, and HG, Eudragit® RSPO, Eudragit® FS100, Kollidon® VA64 and Plasdone™ K-29/32. The amorphous solubility, nucleation induction time, and particle size analysis of nifedipine in a supersaturated solution were performed with and without the presence of polymers, alone or in combination. The HPMCAS-HG and HPMCAS-HG + LG combinations showed the highest nifedipine amorphous solubility of 169.47, 149.151 µg/mL, respectively and delay in nucleation induction time up to 120 min compared to other polymeric combinations. The solid dispersions prepared via hot melt extrusion showed the transformation of crystalline nifedipine to amorphous form. The in-vitro non-sink dissolution study revealed that although the binary nifedipine/HPMCAS-LG system had shown the greater supersaturation concentration of 66.1 µg/mL but could not maintain a supersaturation level up to 360 min. A synergistic effect emerged for ternary nifedipine/HPMCAS-LG/HPMCAS-HG, and nifedipine/HPMCAS-LG/Eudragit®FS100 systems maintained the supersaturation level with enhanced dissolution performance, demonstrating the potential of polymeric combinations for improved amorphous solid dispersion performance.


Sujet(s)
Méthylcellulose , Polymères , Cinétique , Méthylcellulose/analogues et dérivés , Solubilité
14.
Curr Drug Deliv ; 19(1): 32-40, 2022.
Article de Anglais | MEDLINE | ID: mdl-34126896

RÉSUMÉ

BACKGROUND: Exenatide(EXE) is an anti-hyperglycemic agent approved for treating type 2 diabetes by the Food and Drug Administration(FDA). However, twice-daily injection of exenatide is inconvenient for most of the patients. OBJECTIVE: In this study, biotinylated trimethylated chitosan(Bio-TMC) based nanoparticles were proposed to promote oral absorption of exenatide. Realizing the oral administration of exenatide is very important to alleviate patient suffering and improve patient compliance. METHODS: Bio-TMC was synthesized, and the chemical structure was characterized by Fourier transform infrared (FT-IR) spectroscopy and 1H NMR spectroscopy. Nanoparticles were prepared through polyelectrolyte interaction in the presence of sodium Tripolyphosphate (TPP) and hydroxypropyl methylcellulose phthalate (HP-55). Formulations were physically and chemically characterized. In vitro release was investigated in different pH media. In vivo antidiabetic activities of biotin modified and non-biotin modified chitosan were evaluated in db/db mice. RESULTS: EXE-loaded Bio-TMC/HP-55 nanoparticles were spherical in shape with a mean diameter of 156.2 nm and zeta potential of +11.3 mV. The drug loading efficiency and loading content were 52.38% and 2.08%, respectively. In vitro release revealed that EXE-loaded Bio-TMC/HP-55 nanoparticles were released faster in pH 1.2 than pH 6.8 (63.71% VS 50.12%), indicating that nanoparticles have enteric characteristics. Antidiabetic activity study revealed that after oral administration to diabetic mice, the relative pharmacological bioavailability (FPharm%) of the biotin modified nanoparticles was found to be 1.27-fold higher compared to the unmodified ones, and the hypoglycemic effect was also found to be better. CONCLUSION: Bio-TMC/HP-55 nanoparticles are feasible as oral drug carriers of exenatide and have the potential to be extended to other drugs that are not readily oral, such as monoclonal antibodies, vaccines, genes, etc. These would be beneficial to the pharmaceutical industry. Further research will focus on the biodistribution of Bio-TMC/HP-55 nanoparticles after oral administration.


Sujet(s)
Chitosane , Diabète expérimental , Diabète de type 2 , Nanoparticules , Animaux , Chitosane/composition chimique , Diabète expérimental/traitement médicamenteux , Vecteurs de médicaments/composition chimique , Exénatide , Humains , Méthylcellulose/analogues et dérivés , Souris , Nanoparticules/composition chimique , Taille de particule , Spectroscopie infrarouge à transformée de Fourier , Distribution tissulaire
15.
Mol Pharm ; 19(1): 51-66, 2022 01 03.
Article de Anglais | MEDLINE | ID: mdl-34919407

RÉSUMÉ

Understanding the dissolution mechanisms of amorphous solid dispersions (ASDs) and being able to link enhanced drug exposure with process parameters are key when formulating poorly soluble compounds. Thus, in this study, ASDs composed by itraconazole (ITZ) and hydroxypropylmethylcellulose acetate succinate (HPMCAS) were formulated with different polymer grades and drug loads (DLs) and processed by spray drying with different atomization ratios and outlet temperatures. Their in vitro performance and the ability to form drug-rich colloids were then evaluated by a physiologically relevant dissolution method. In gastric media, drug release followed a diffusion-controlled mechanism and drug-rich colloids were not formed since the solubility of the amorphous API at pH 1.6 was not exceeded. After changing to intestinal media, the API followed a polymer dissolution-controlled release, where the polymer rapidly dissolved, promoting the immediate release of API and thus leading to liquid-liquid phase separation (LLPS) and consequent formation of drug-rich colloids. However, the release of API and polymer was not congruent, so API surface enrichment occurred, which limited the further dissolution of the polymer, leading to a drug-controlled release. ASDs formulated with M-grade showed the highest ability to maintain supersaturation and the lowest tendency for AAPS due to its good balance between acetyl and succinoyl groups, and thus strong interactions with both the hydrophobic drug and the aqueous dissolution medium. The ability to form colloids increased for low DL (15%) and high specific surface area due to the high amount of polymer released until the occurrence of API surface enrichment. Even though congruent release was not observed, all ASDs formed drug-rich colloids that were stable in the solution until the end of the dissolution study (4 h), maintaining the same size distribution (ca. 300 nm). Drug-rich colloids can, in vivo, act as a drug reservoir replenishing the drug while it permeates. Designing ASDs that are prone to form colloids can overcome the solubility constraints of Biopharmaceutics Classification System (BCS) II and IV drugs, posing as a reliable formulation strategy.


Sujet(s)
Colloïdes/composition chimique , Préparation de médicament , Itraconazole/administration et posologie , Méthylcellulose/analogues et dérivés , Calorimétrie différentielle à balayage , Association médicamenteuse , Préparation de médicament/méthodes , Libération de médicament , Itraconazole/analyse , Itraconazole/composition chimique , Méthylcellulose/administration et posologie , Méthylcellulose/analyse , Méthylcellulose/composition chimique , Microscopie électronique à balayage , Taille de particule , Diffraction des rayons X
16.
Pharm Res ; 38(12): 2119-2127, 2021 Dec.
Article de Anglais | MEDLINE | ID: mdl-34931285

RÉSUMÉ

PURPOSE: The intestinal fluid pH is maintained by the bicarbonate buffer system that shows unique properties regarding drug dissolution. Nevertheless, current compendial dissolution tests use phosphate buffers. The purpose of the present study was to investigate the effect of bicarbonate and phosphate buffers on the dissolution profiles of amorphous solid dispersions (ASD) composed of ionizable polymers. METHODS: Hydroxypropylmethylcellulose acetate succinate (HPMCAS), amino methacrylate copolymer (AMC), and hydroxypropylmethylcellulose (HPMC) were employed as acidic, basic, and neutral polymers, respectively. Nifedipine (NIF) was used as a model drug. Dissolution profiles were measured in pH 6.5 bicarbonate and phosphate buffers by a mini-scale paddle dissolution test. The pH of bicarbonate buffers was maintained by the floating lid method. RESULTS: The pH change of the bicarbonate buffer was suppressed to less than + 0.25 pH for 3 h by the floating lid method. In all cases, the NIF concentration was supersaturated against the solubility of crystalline NIF. The dissolution rates of HPMCAS and AMC ASDs were 1.5 to 2.0-fold slower in the bicarbonate buffer than in the phosphate buffer when compared at the same buffer capacity. The dissolution profile of HPMC ASD was not affected by the buffer species. The higher the buffer capacity and ionic strength, the faster the dissolution rate of HPMCAS ASD. CONCLUSION: The dissolution rate of ASDs with ionizable polymers would be overestimated by using unphysiological phosphate buffer solutions. It is important to use a biorelevant bicarbonate buffer solution for dissolution testing.


Sujet(s)
Vecteurs de médicaments/composition chimique , Nifédipine/pharmacocinétique , Hydrogénocarbonates/composition chimique , Substances tampon , Chimie pharmaceutique , Libération de médicament , Concentration en ions d'hydrogène , Méthylcellulose/analogues et dérivés , Méthylcellulose/composition chimique , Nifédipine/administration et posologie , Phosphates/composition chimique , Polymères/composition chimique , Solubilité
17.
Mol Pharm ; 18(12): 4299-4309, 2021 12 06.
Article de Anglais | MEDLINE | ID: mdl-34738825

RÉSUMÉ

Polymers play an important role in amorphous solid dispersions (ASDs), enhancing stability in the solid state and maintaining supersaturation in aqueous solutions of intrinsically low-water-soluble drug candidates. Hydroxypropyl methylcellulose acetate succinate (HPMCAS) is widely used in ASDs due to its hydrophobic/hydrophilic balance and ionizability of the substituent functionalities. While colloid formation of HPMCAS in solution due to this hydrophobic/hydrophilic balance has been studied, the impact of the polymer conformation (random coil vs aggregated) on drug supersaturation of ASDs is not well understood. To our knowledge, this is the first report where the critical aggregation concentration for three grades of HPMCAS (HF/MF/LF) has been determined via fluorescence spectroscopy using the environment-sensitive probe pyrene. The specific impact of polymer conformation (random coil vs aggregate) on the model drug celecoxib (CLX) has been elucidated with fluorescence quenching and nuclear magnetic resonance (NMR) spectroscopy. A negative deviation of the Stern-Volmer plot indicated that aggregated HPMCAS effectively blocked the quencher's access to CLX. This is further supported by NMR observations, where NMR spectra indicate a larger change of chemical shift of the -NH group of CLX when HPMCAS is above its aggregated concentration, suggesting strong H-bonding interactions between aggregated HPMCAS and CLX. Finally, the supersaturation-precipitation study shows that all three grades of HPMCAS in the aggregated state significantly enhanced CLX supersaturation compared to the nonaggregated state, indicating that polymer aggregation plays a critical role in maintaining drug supersaturation.


Sujet(s)
Célécoxib/composition chimique , Méthylcellulose/analogues et dérivés , Précipitation chimique , Cristallisation , Spectroscopie par résonance magnétique , Méthylcellulose/composition chimique , Solubilité , Spectrométrie de fluorescence
18.
Eur J Pharm Biopharm ; 169: 189-199, 2021 Dec.
Article de Anglais | MEDLINE | ID: mdl-34756974

RÉSUMÉ

Poor solubility of drug candidates is a well-known and thoroughly studied challenge in the development of oral dosage forms. One important approach to tackle this challenge is the formulation as an amorphous solid dispersion (ASD). To reach the desired biopharmaceutical improvement a high supersaturation has to be reached quickly and then be conserved long enough for absorption to take place. In the presented study, various formulations of regorafenib have been produced and characterized in biorelevant in-vitro experiments. Povidone-based formulations, which are equivalent to the marketed product Stivarga®, showed a fast drug release but limited stability and robustness after that. In contrast, HPMCAS-based formulations exhibited excellent stability of the supersaturated solution, but unacceptably slow drug release. The attempt to combine the desired attributes of both formulations by producing a ternary ASD failed. Only co-administration of HPMCAS as an external stabilizer to the rapidly releasing Povidone-based ASDs led to the desired dissolution profile and high robustness. This optimized formulation was tested in a pharmacokinetic animal model using Wistar rats. Despite the promising in-vitro results, the new formulation did not perform better in the animal model. No differences in AUC could be detected when compared to the conventional (marketed) formulation. These data represent to first in-vivo study of the new concept of external stabilization of ASDs. Subsequent in-vitro studies revealed that temporary exposure of the ASD to gastric medium had a significant and long-lasting effect on the dissolution performance and externally administered stabilizer could not prevent this sufficiently. By applying the co-administered HPMCAS as an enteric coating onto Stivarga tablets, a new bi-functional approach was realized. This approach achieved the desired tailoring of the dissolution profile and high robustness against gastric medium as well as against seeding.


Sujet(s)
Libération de médicament/effets des médicaments et des substances chimiques , Méthylcellulose/analogues et dérivés , Phénylurées , Pyridines , Solubilité/effets des médicaments et des substances chimiques , Animaux , Produits biologiques/administration et posologie , Produits biologiques/pharmacocinétique , Formes posologiques , Voies d'administration de substances chimiques et des médicaments , Préparation de médicament/méthodes , Excipients/administration et posologie , Excipients/pharmacocinétique , Méthylcellulose/administration et posologie , Méthylcellulose/pharmacocinétique , Phénylurées/administration et posologie , Phénylurées/pharmacocinétique , Povidone/composition chimique , Povidone/pharmacologie , Pyridines/administration et posologie , Pyridines/pharmacocinétique , Rats , Extraction en phase solide/méthodes , Comprimés entérosolubles/administration et posologie , Comprimés entérosolubles/pharmacocinétique
19.
Int J Biol Macromol ; 190: 989-998, 2021 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-34537299

RÉSUMÉ

The present study aimed to investigate the thermal- and pH-dependent gelation behavior of gelatin/HPMCP blends using ultraviolet (UV) spectrophotometry, viscoelasticity, and dynamic light scattering (DLS). We found that the release of lisinopril from gelatin/HPMCP gels can be inhibited at low pH. UV spectrophotometric analysis showed that pH had a significant effect on the transparency of aqueous HPMCP systems and gelatin/HPMCP gels. The viscoelastic patterns of gelatin/HPMCP at pH 4.6 considerably differed from those of gelatin/HPMCP at pH 5.2 and 6.0. DLS measurements showed that HPMCP molecules in low concentrations underwent strong aggregation below pH 4.8. Such HPMCP aggregation induces a physical barrier in the matrix structures of the gelatin/HPMCP gels, which inhibits the drug release at pH 1.2. This hydrogel delivery system using polymer blends of gelatin/HPMCP can be used in oral gel formulations with pH-responsive properties.


Sujet(s)
Libération de médicament , Gélatine/composition chimique , Méthylcellulose/analogues et dérivés , Diffusion dynamique de la lumière , Module d'élasticité , Concentration en ions d'hydrogène , Lisinopril/pharmacologie , Méthylcellulose/composition chimique , Polymères/composition chimique , Rhéologie , Électricité statique
20.
Pak J Pharm Sci ; 34(1(Supplementary)): 225-235, 2021 Jan.
Article de Anglais | MEDLINE | ID: mdl-34275846

RÉSUMÉ

The objective was to develop eperisone HCl sustained-release pellets through extrusion spheronization technique and to determine the influence of different hydrophobic (polymeric based and wax-based) and hydrophilic (polymeric based) matrix former on the release of eperisone HCl (BCS class I drug) and on pellet sphericity. The pellet formulations consisted of different hydrophobic and hydrophilic matrix formers like HPMC K4M (10-20%) HPMC K15M (10%), EC (7cps) (10-20%), Carnauba wax (10-20%), Compritol ATO 888 (10-20%), Glyceryl monostearate (10%), lactose and microcrystalline cellulose. The initial burst release of the drug from matrix pellet formulations was effectively controlled by coating with 5% EC (ethylcellulose) dispersion. The dissolution profile and drug release kinetics of coated pellet formulations were determined at both acidic and basic pH medium. SEM (Scanning electron microscope) technique was used to determine the surface morphology and cross-section of F5 and F7 pellet formulation. The mechanism of drug release of coated formulation followed non-Fickian diffusion. FTIR spectroscopy was conducted and no drug and excipients interaction was observed. The results had shown that optimized coated formulation was F5 and F7 which effectively extend the drug release for 12 hours.


Sujet(s)
Préparations à action retardée/pharmacocinétique , Myorelaxants à action centrale/pharmacocinétique , Propiophénones/pharmacocinétique , Cellulose/analogues et dérivés , Chimie pharmaceutique , Préparations à action retardée/composition chimique , Développement de médicament , Libération de médicament , Excipients/composition chimique , Acides gras , Glycérides , Lactose/analogues et dérivés , Méthylcellulose/analogues et dérivés , Microscopie électronique à balayage , Myorelaxants à action centrale/administration et posologie , Myorelaxants à action centrale/composition chimique , Polymères , Propiophénones/administration et posologie , Propiophénones/composition chimique , Spectroscopie infrarouge à transformée de Fourier , Cires
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...