Jin-Hyuk Jeong, Chang-Soo Han, Ji-Hyun Kang, Kwang-Hwi Yoo, Woong-Young Jung, Yun-Sang Park, Dong-Wook Kim, Chun-Woong Park
{"title":"Preparation and characterization of immediate release 3D printed tablets using hot melt extruded amorphous cyclosporine a filament.","authors":"Jin-Hyuk Jeong, Chang-Soo Han, Ji-Hyun Kang, Kwang-Hwi Yoo, Woong-Young Jung, Yun-Sang Park, Dong-Wook Kim, Chun-Woong Park","doi":"10.1080/10837450.2025.2472893","DOIUrl":"10.1080/10837450.2025.2472893","url":null,"abstract":"<p><p>3D printing technology is gaining attention as a next-generation approach to drug formulation. Among 3D printing techniques, fused deposition modeling is cost-effective but depends heavily on suitable filaments. Hot melt extrusion enables filament production by incorporating poorly water-soluble drugs like cyclosporine A into polymers to form solid dispersions. However, achieving immediate release formulations with 3D printing remains challenging due to issues such as inadequate tablet disintegration or drug entrapment within the polymer matrix. This study aimed to develop and evaluate immediate release 3D-printed cyclosporine A tablets using HME filaments. Three parameters were modified in the 3D printing process: varying fill speeds, infill densities, and channel lengths. Filaments composed of Kollidon<sup>®</sup> VA 64 and HPC-SSL (1:1) were used to print tablets. Solid-state analysis confirmed cyclosporine A 's amorphous state and partial crystallinity in Xylisorb<sup>®</sup> 300. Dissolution studies revealed that lower infill densities (30%) and fewer walls enhanced drug release by increasing internal void space and reducing hardness. Conversely, greater tablet height (channel length) delayed dissolution. These findings emphasize the critical role of geometric design in drug release, showcasing the potential of 3D printing to create personalized dosage forms tailored to patient needs by optimizing structural parameters.</p>","PeriodicalId":20004,"journal":{"name":"Pharmaceutical Development and Technology","volume":" ","pages":"1-11"},"PeriodicalIF":2.6,"publicationDate":"2025-03-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143537530","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"<i>Saccharomyces cerevisiae</i>-derived vesicles loaded with dextromethorphan as a candidate for the management of neuroinflammation related to Alzheimer's disease.","authors":"Parastoo Valizadeh, Negin Mozafari, Hajar Ashrafi, Reza Heidari, Negar Azarpira, Amir Azadi","doi":"10.1080/10837450.2025.2470351","DOIUrl":"10.1080/10837450.2025.2470351","url":null,"abstract":"<p><p>Alzheimer's disease (AD) is a neurodegenerative disease that is associated with neuroinflammation. Dextromethorphan (DXM) exerts neuroprotective effects in many central nervous system injuries and neurodegenerative diseases. The cell wall of <i>Saccharomyces cerevisiae</i> is a cell-based drug delivery system that can be a suitable candidate for targeted drug delivery to the site of inflammation. In this study, nanoparticles (NPs) were prepared from <i>Saccharomyces cerevisiae</i> cell walls, coated with polysorbate-80, and loaded with DXM. NPs had favorable hemolytic behavior with a particle size distribution of 187.25 ± 73.37 nm and a zeta potential of +4.3 mV. Pathological examination in a mouse model of neuroinflammation showed that NPs had the ability to reduce brain inflammation and the adverse effects of DXM.</p>","PeriodicalId":20004,"journal":{"name":"Pharmaceutical Development and Technology","volume":" ","pages":"1-9"},"PeriodicalIF":2.6,"publicationDate":"2025-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143524119","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Preparation of diltiazem HCl-modified release formulation using cation-exchange resin as a single excipient.","authors":"Khouloud A Alkhamis, Suhair S Al-Nimry","doi":"10.1080/10837450.2025.2474092","DOIUrl":"10.1080/10837450.2025.2474092","url":null,"abstract":"<p><strong>Objective: </strong>Modified released formulations of diltiazem were previously prepared using cation-exchange resins. However, multiple excipients were required to achieve the appropriate release rate. It was of interest to prepare a modified release dosage form of diltiazem using drug-resin complex alone.</p><p><strong>Methods: </strong>Adsorption experiments conducted using a rotating bottle apparatus. The procedure involved adding the resin to the bottles, followed by appropriate amount of drug solution. The bottles were rotated until equilibrium was reached and the concentrations were analyzed using a reversed phase HPLC method, which effectively separated the compound from its degradation product. Release studies were conducted using a USP dissolution apparatus 2 with phosphate buffer as the dissolution medium.</p><p><strong>Key findings: </strong>Diltiazem was unstable inside the resin when the H<sup>+</sup> form was used. It became stable when the H<sup>+</sup> was displaced with Na<sup>+</sup>. Langmuir-like equation was applied to the adsorption isotherms. The equation parameters were influenced by the resin's cross-linking and particle size. Maximum drug release is related to sample volume. Positive linear relationship was obtained between initial release rate and extent of uptake.</p><p><strong>Conclusion: </strong>This study successfully demonstrates that Dowex® 50WX8 (Na<sup>+</sup> form) can be used as a single excipient in diltiazem formulations, providing both chemical stability and sustained release without requiring additional polymer coatings.</p>","PeriodicalId":20004,"journal":{"name":"Pharmaceutical Development and Technology","volume":" ","pages":"1-8"},"PeriodicalIF":2.6,"publicationDate":"2025-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143524120","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Shahinaze A Fouad, Nada Abdelaziz, Mahmoud H Teaima, Mohamed El-Nabarawi, Amal Anwar Taha, Rehab Abdelmonem, Khaled El-Refai
{"title":"Engineering orally disintegrating tablets for buccal delivery of cilostazol with enhanced dissolution and bioavailability: a novel dual porogenic approach, <i>in vitro</i> characterization, and <i>in vivo</i> evaluation in rats.","authors":"Shahinaze A Fouad, Nada Abdelaziz, Mahmoud H Teaima, Mohamed El-Nabarawi, Amal Anwar Taha, Rehab Abdelmonem, Khaled El-Refai","doi":"10.1080/10837450.2025.2472887","DOIUrl":"10.1080/10837450.2025.2472887","url":null,"abstract":"<p><p>Cilostazol (CTZ), is a BCS class II drug with limited bioavailability. In the current study, orally disintegrating tablets (ODTs) for buccal delivery of CTZ were prepared by two methods; lyophilization (Lyo-ODTs) and direct compression (DC-ODTs). All CTZ-ODTs were evaluated for <i>in vitro</i> disintegration time (DT) and wetting time (WT) tests, <i>in vitro</i> dissolution. Scanning electron microscopic (SEM) analysis was performed for the selected Lyo-ODT-7 and DC-ODT-2. Lyo-ODT-7 composed of aerosil<sup>®</sup> 200 and PEG 4000 acquired the shortest DT (13.00 ± 0.14) and WT (33.00 ± 0.26) among the prepared ODTs. It also showed a 2.3 fold significantly enhanced dissolution profile at an early time point (5 min) that was maintained till 1 h, in simulated saliva fluid (pH ∼ 6.8), compared to Pletaal<sup>®</sup> IR tablets (<i>p</i> < 0.0001). SEM analysis revealed the remarkable porosity of Lyo-ODT-7, confirming its successfully enhanced disintegration and dissolution. Lyo-ODT-7 showed significantly enhanced pharmacokinetic parameters with a 3.5 and 3.6 fold increase in C<sub>max</sub> (<i>p</i> = 0.0493) and AUC<sub>0-24</sub> (<i>p</i> = 0.0470), respectively compared to Pletaal<sup>®</sup> IR tablets. The relative bioavailability of CTZ after buccal administration of Lyo-ODT-7 to rats was 364.45%, compared to the market oral IR tablets; Pletaal<sup>®</sup>. The enhanced bioavailability imposes the successful oromucosal absorption of CTZ <i>via</i> buccal delivery of Lyo-ODT-7. Our study demonstrated that Lyo-ODT-7 could represent a favorable buccal dosage form for patients with intermittent claudication, suffering from dysphagia. It can also be used in cases of acute cerebral or myocardial infarction due to its significantly enhanced rate and extent of absorption. It is considered a promising approach for buccal delivery of BCS class II active pharmaceutical ingredients (APIs) suffering from solubility problems and hepatic first pass effect.HIGHLIGHTSODTs containing CTZ were prepared via lyophilization and direct compression to produce Lyo-ODTs and DC-ODTs, respectively.Lyo-ODT-7 displayed the fastest DT, shortest WT and exhibited significantly enhanced in vitro dissolution at 5 minutes compared to Pletaal<sup>®</sup> IR tablets (<i>p</i> < 0.0001), at the buccal pH (6.8).SEM images of Lyo-ODT-7 elucidated its extremely porous structure with the appearance of wide pores having average pore diameter that is 2.27 fold significantly greater than the directly compressible ODT-2 (DC-ODT-2) (<i>p</i> = 0.0286).Lyo-ODT-7 also showed significantly enhanced C<sub>max</sub> (<i>p</i> = 0.0493) and AUC<sub>0-24</sub> (<i>p</i> = 0.0470) that were 3.5 and 3.6 fold greater than Pletaal<sup>®</sup> IR tablets, respectively with improved relative bioavailability ∼ 364.45%.A dual porogenic effect was achieved via implementing both lyophilization technique and a pore-former excipient, during pharmaceutical development of CTZ-loaded ODT (Lyo-ODT-7), and ","PeriodicalId":20004,"journal":{"name":"Pharmaceutical Development and Technology","volume":" ","pages":"1-15"},"PeriodicalIF":2.6,"publicationDate":"2025-02-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143503322","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Ezetimibe Oral Solid Lipid Nanoparticle by Effervescent Dispersion Method: <i>in Vitro</i> Characterization And <i>in Vivo</i> Pharmacokinetic Study in Rats.","authors":"Ruba S Darweesh, Farah S AlQawasmi, Mai S Khanfar","doi":"10.1080/10837450.2025.2471461","DOIUrl":"https://doi.org/10.1080/10837450.2025.2471461","url":null,"abstract":"<p><p>Ezetimibe (EZT) is a class II drug of the Biopharmaceutics classification system (BCS), with limited aqueous solubility and high permeability. This study aims to enhance the solubility and oral bioavailability of EZT by developing EZT solid lipid nanoparticles (SLNs). EZT-SLNs were developed through the effervescent dispersion technique. Different amounts of Tween-80, Compritol ATO 888, and mannitol as cryoprotectant were used. F11 was the optimum formula with 154 nm in size and 90.26% entrapment efficiency. It demonstrates significant enhancements in solubility across various pH values. In addition, F11 shows a significantly higher drug release than pure EZT at all time points, and that's related to the reduction in the particle size and increasing its surface area along with the transformation from a crystalline state to an amorphous state. The powder X-ray diffraction and Differential Scanning Calorimetry tests confirmed this conversion from crystalline form to amorphous. The <i>in vivo</i> animal study demonstrated that the C<sub>max</sub> and <math><msubsup><mrow><mtext> AUC</mtext></mrow><mrow><mn>0</mn></mrow><mrow><mtext>∞ </mtext></mrow></msubsup></math> of the EZT-SLNs group were significantly higher than the pure EZT group, after oral administration. In conclusion, EZT-SLNs with enhanced <i>in vitro</i> and <i>in vivo</i> properties were successfully developed using the effervescent dispersion technique.</p>","PeriodicalId":20004,"journal":{"name":"Pharmaceutical Development and Technology","volume":" ","pages":"1-36"},"PeriodicalIF":2.6,"publicationDate":"2025-02-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143483886","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Ocular polymeric nanomicelles for the posterior eye segment in the management of retinoblastoma: formulation, optimization, <i>in vitro</i> and <i>ex vivo</i> evaluations.","authors":"Mudassir Ansari, Yogesh A Kulkarni, Kavita Singh","doi":"10.1080/10837450.2025.2469321","DOIUrl":"10.1080/10837450.2025.2469321","url":null,"abstract":"<p><p>The existing study focuses on the development, optimization, and evaluation of sorafenib-loaded polymeric nanomicelles for posterior segment delivery in treating retinoblastoma. The formulation involved adjusting various process and product parameters to create effective drug-loaded polymeric nanomicelles. The particle size, PDI, and zeta potential of optimized formulation were found to be 65.52 ± 1.18 nm, 0.14 ± 0.01, and -3.26 ± 0.66 mV, respectively. The entrapment efficiency and drug release were estimated to be 98.84% ± 0.001 and 99.99% in 6 h, respectively. Additionally, the optimized formulation demonstrated acceptable outcomes for solid-state analysis, osmolality, pH, residual solvent, and morphological properties. After 8 h, the <i>ex vivo</i> transcleral permeation and scleral deposition were 629.05 ± 124.11 ng/cm<sup>2</sup> and 4.10 ± 0.54 µg, respectively. Y-79 (human retinoblastoma) cell line study using standard drug, test drug, and optimized formulation revealed anticancer potential at all time points (6, 12, 18, and 24 h) with comparable IC50 values. Furthermore, the optimized formulation exhibited no toxicity on the ARPE-19 (human retinal pigmented epithelium) cell line over 24 h. The optimized formulation was non-irritating to the eye (HET-CAM) and remained stable for 6 months. Thus, drug delivery to the posterior eye segment for the treatment of retinoblastoma appears to be possible with the help of established technology.</p>","PeriodicalId":20004,"journal":{"name":"Pharmaceutical Development and Technology","volume":" ","pages":"1-13"},"PeriodicalIF":2.6,"publicationDate":"2025-02-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143476825","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Formulation and optimization of glycyrrhetinic acid-modified pH-sensitive curcumin liposomes for anti-hepatocellular carcinoma.","authors":"Jie Wang, Xuemei Gu, Xia Gao, Jing Chen, Zhiyang Lv, Siyu Zhang, Siyu Ni, Fei Shi, Xialin Chen, Liang Cao, Zhenzhong Wang, Wei Xiao","doi":"10.1080/10837450.2025.2465549","DOIUrl":"10.1080/10837450.2025.2465549","url":null,"abstract":"<p><p>In order to enhance the therapeutic value of curcumin in liver cancer treatment, glycyrrhetinic acid-modified pH-sensitive curcumin liposomes (GA-pH-Lip@Cur) was developed.GA-pH-Lip@Cur was prepared using a thin film dispersion ultrasonication method, and the optimal formulation process was selected through single-factor experiments and a Box-Behnken design-response surface methodology. The liposomes were evaluated for their morphological appearance, particle size, <i>in vitro</i> release at different pH levels, and biocompatibility. The anti-tumor effect of GA-pH-Lip@Cur was assessed using cell viability assays (CCK-8). The <i>in vivo</i> hepatic targeting and anti-liver tumor efficacy of GA-pH-Lip@Cur were evaluated through pharmacokinetic and pharmacological experiments.The results indicated that optimized GA-pH-Lip@Cur exhibited uniform particle size distribution, good stability, pH-sensitive <i>in vitro</i> release with sustained behavior. Compared to conventional liposomes, GA-pH-Lip@Cur showed prolonged average retention time <i>in vivo</i> and significantly increased curcumin distribution in liver tissues, indicating excellent liver targeting. Both <i>in vitro</i> and <i>in vivo</i> evaluations demonstrated the effectiveness of GA-pH-Lip@Cur in inhibiting liver cancer cell proliferation and suppressing liver tumor growth in tumor-bearing mice. In conclusion, GA-pH-Lip@Cur, by leveraging the acidic tumor microenvironment and overexpression of glycyrrhetinic acid receptors in liver cells, encapsulates curcumin to improve its bioavailability, and target its delivery to the liver tumor sites.</p>","PeriodicalId":20004,"journal":{"name":"Pharmaceutical Development and Technology","volume":" ","pages":"1-13"},"PeriodicalIF":2.6,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143399752","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jiayu Xie, Ke Li, Lusi Chen, Huiying Zhong, Tao Xiao, Lihua Chen, Haibing He, Hongfei Liu, Guoqing Zhang
{"title":"Long-acting release of fluocinolone acetonide microspheres using electrospray technology for noninfectious uveitis therapy.","authors":"Jiayu Xie, Ke Li, Lusi Chen, Huiying Zhong, Tao Xiao, Lihua Chen, Haibing He, Hongfei Liu, Guoqing Zhang","doi":"10.1080/10837450.2025.2462998","DOIUrl":"10.1080/10837450.2025.2462998","url":null,"abstract":"<p><p>Intravitreous long-acting drug delivery system offers an excellent alternative to multiple injections for the treatment of noninfectious uveitis (NIU). However, the adverse effects of non-biodegradable intravitreal implants of fluocinolone acetonide (FA), such as postoperative hypotony and secondary injury during removal of the implant matrix, are frequent occurrence to affect patient's compliance. Herein, biodegradable poly (lactic-co-glycolic acid) (PLGA)-based microspheres (MS) containing fluocinolone acetonide (FA@MS) were prepared using an optimized electrospray technology with a voltage of 10.07 kV and the receiving distance of 9.87 cm. The obtained FA@MS with the average particle size of 2.25 μm possessed the high encapsulation efficiency (94.85%) and drug content (9.48%). <i>In vitro</i> release demonstrated that FA@MS exhibited sustained release for 30 days, and the release characteristic of FA@MS conformed to the Weibull model. <i>In vivo</i> study in a rabbit NIU model indicated that FA@MS continuously released the drug for at least 28 days in vitreum and progressively decreased inflammation of NIU. Furthermore, the intraocular pressure of rabbits treated with blank MS and FA@MS remained the normal level for 28 days, which demonstrated the favorable biosafety of FA@MS. In conclusion, long-acting release of FA@MS provides a promising formulation for NIU treatment. HIGHLIGHTSA biodegradable FA@MS was prepared using the modified electrospray technology for intravitreal administration.FA@MS exhibited the sustained release characteristics for 30 days in the medium of PBS (pH 7.4) with 0.2% Tween 80.The pharmacodynamics indicated that FA@MS could be continuously released for at least 28 days in vitreum to treat NIU.</p>","PeriodicalId":20004,"journal":{"name":"Pharmaceutical Development and Technology","volume":" ","pages":"210-219"},"PeriodicalIF":2.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143123412","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"NGR-poly(2-ethyl-2-oxazoline)-cholesteryl methyl carbonate enhances the antitumor effect of quercetin liposomes in triple-negative breast cancer.","authors":"Chengcheng Zhao, Jian Qin, Dingyu Zhang, Xue Li, Ning Yang, Tingyu Gao, Junliang Song, Yule Song, Shouzhen Huang, Huan Xu","doi":"10.1080/10837450.2025.2450434","DOIUrl":"10.1080/10837450.2025.2450434","url":null,"abstract":"<p><p>In this paper, the pH-sensitive targeting functional material NGR-poly(2-ethyl-2-oxazoline)-cholesteryl methyl carbonate (NGR-PEtOz-CHMC, NPC) modified quercetin (QUE) liposomes (NPC-QUE-L) was constructed. The structure of NPC was confirmed by infrared spectroscopy (IR) and nuclear magnetic resonance hydrogen spectrum (<sup>1</sup>H-NMR). Pharmacokinetic results showed that the accumulation of QUE in plasma of the NPC-QUE-L group was 1.28 times and 2.43 times that of the QUE Solution and QUE-L groups, respectively. The release amount of NPC-QUE-L in an acidic environment was significantly higher than in physiological pH value. The order of the tumor cell inhibition rate in different pH environments was NPC-QUE-L > PC-QUE-L > QUE-L. In addition, the cellular uptake of NPC-modified liposomes was higher than that of PC-modified and unmodified liposomes, indicating that NPC had good pH-sensitivity and targeting. In the triple-negative breast cancer (TNBC) model, the relative tumor proliferation rate of NPC-QUE-L is about 73%, which is better than that of the QUE solution group. Western blot results show that NPC-QUE-L can effectively reduce the expression of α-smooth actin and transforming growth factor-β1 in tumor tissues, and improve the degree of tumor fibrosis. In this study, NPC could endow QUE liposomes with good stability, pH-sensitivity, and targeting, which provides a reference for improving the solubility and targeting of poorly soluble natural drug components.</p>","PeriodicalId":20004,"journal":{"name":"Pharmaceutical Development and Technology","volume":" ","pages":"137-149"},"PeriodicalIF":2.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143009696","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Formulation development and evaluation of push-pull osmotic pump bi-layered tablets for phencynonate HCl in the treatment of motion sickness.","authors":"Si-Rui Liu, Wen Lin, Xiang-Yang Xie, Yuan Zeng","doi":"10.1080/10837450.2025.2465548","DOIUrl":"10.1080/10837450.2025.2465548","url":null,"abstract":"<p><p>The aim of this study was to develop and evaluate an extended-release (ER) push-pull osmotic pump (PPOP) tablet for phencynonate HCl (PCN), which could release the drug at zero-order profile for a duration of 24 h. The core tablets were designed as bi-layered, primarily composed of sodium chloride and polyethylene oxide (PEO). The central composite design (CCD) within a response surface methodology (RSM) was used to optimize the formulation. An optimized PCN-PPOP tablet formulation was achieved with the following values for key factors: 10 mg NaCl, 70 mg PEO, and 13.56% coating membrane weight gain. It revealed that this formulation could release PCN <i>in vitro</i> at a zero-order manner for 18 h. The <i>in vivo</i> release property of the PCN-PPOP tablet was assessed and contrasted with that of immediate-release (IR) tablet following a single oral administration to beagle dogs. The pharmacokinetic data indicated that the PPOP tablet achieved a sustained <i>in vivo</i> release of PCN, as evidenced by a longer T<sub>max</sub> (7.17 ± 1.83 h) and mean residence time (11.57 ± 1.12 h). This work demonstrated that PCN-PPOP tablet could be designed for oral administration to provide a long-term pharmacological intervention for motion sickness.</p>","PeriodicalId":20004,"journal":{"name":"Pharmaceutical Development and Technology","volume":" ","pages":"220-232"},"PeriodicalIF":2.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143391553","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}