1
|
Real JP, Real DA, Lopez-Vidal L, Barrientos BA, Bolaños K, Tinti MG, Litterio NJ, Kogan MJ, Palma SD. 3D-Printed Gastroretentive Tablets Loaded with Niclosamide Nanocrystals by the Melting Solidification Printing Process (MESO-PP). Pharmaceutics 2023; 15:pharmaceutics15051387. [PMID: 37242629 DOI: 10.3390/pharmaceutics15051387] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/13/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Niclosamide (NICLO) is a recognized antiparasitic drug being repositioned for Helicobacter pylori. The present work aimed to formulate NICLO nanocrystals (NICLO-NCRs) to produce a higher dissolution rate of the active ingredient and to incorporate these nanosystems into a floating solid dosage form to release them into the stomach slowly. For this purpose, NICLO-NCRs were produced by wet-milling and included in a floating Gelucire l3D printed tablet by semi-solid extrusion, applying the Melting solidification printing process (MESO-PP) methodology. The results obtained in TGA, DSC, XRD and FT-IR analysis showed no physicochemical interactions or modifications in the crystallinity of NICLO-NCR after inclusion in Gelucire 50/13 ink. This method allowed the incorporation of NICLO-NCRs in a concentration of up to 25% w/w. It achieved a controlled release of NCRs in a simulated gastric medium. Moreover, the presence of NICLO-NCRs after redispersion of the printlets was observed by STEM. Additionally, no effects on the cell viability of the NCRs were demonstrated in the GES-1 cell line. Finally, gastroretention was demonstrated for 180 min in dogs. These findings show the potential of the MESO-PP technique in obtaining slow-release gastro-retentive oral solid dosage forms loaded with nanocrystals of a poorly soluble drug, an ideal system for treating gastric pathologies such as H. pylori.
Collapse
Affiliation(s)
- Juan Pablo Real
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), CONICET, Haya de la Torre y Medina Allemde, Córdoba X5000HUA, Argentina
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Córdoba X5000XHUA, Argentina
| | - Daniel Andrés Real
- Departamento de Química Farmacológica y Toxicológica, Universidad de Chile, Santos Dumont 964, Santiago 8380494, Chile
- Advanced Center of Chronic Diseases (ACCDiS), Universidad de Chile, Santos Dumont 964, Santiago 8380494, Chile
| | - Lucía Lopez-Vidal
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), CONICET, Haya de la Torre y Medina Allemde, Córdoba X5000HUA, Argentina
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Córdoba X5000XHUA, Argentina
| | - Bruno Andrés Barrientos
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), CONICET, Haya de la Torre y Medina Allemde, Córdoba X5000HUA, Argentina
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Córdoba X5000XHUA, Argentina
| | - Karen Bolaños
- Departamento de Química Farmacológica y Toxicológica, Universidad de Chile, Santos Dumont 964, Santiago 8380494, Chile
- Advanced Center of Chronic Diseases (ACCDiS), Universidad de Chile, Santos Dumont 964, Santiago 8380494, Chile
- Center for Studies on Exercise, Metabolism and Cancer (CEMC), Laboratory of Cellular Communication, Program of Cell and Molecular Biology, Faculty of Medicine, Institute of Biomedical Sciences (ICBM), Universidad de Chile, Santiago 8380453, Chile
| | - Mariano Guillermo Tinti
- Facultad de Ciencias Agropecuarias, IRNASUS CONICET, Universidad Católica de Córdoba, Córdoba X5016DHK, Argentina
| | - Nicolás Javier Litterio
- Facultad de Ciencias Agropecuarias, IRNASUS CONICET, Universidad Católica de Córdoba, Córdoba X5016DHK, Argentina
| | - Marcelo Javier Kogan
- Departamento de Química Farmacológica y Toxicológica, Universidad de Chile, Santos Dumont 964, Santiago 8380494, Chile
- Advanced Center of Chronic Diseases (ACCDiS), Universidad de Chile, Santos Dumont 964, Santiago 8380494, Chile
| | - Santiago Daniel Palma
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), CONICET, Haya de la Torre y Medina Allemde, Córdoba X5000HUA, Argentina
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, Córdoba X5000XHUA, Argentina
| |
Collapse
|
2
|
Palekar S, Mamidi HK, Guo Y, Vartak R, Patel K. Corroborating various material-sparing techniques with hot melt extrusion for the preparation of triclabendazole amorphous solid dispersions. Int J Pharm 2023; 640:122989. [PMID: 37120123 DOI: 10.1016/j.ijpharm.2023.122989] [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: 11/04/2022] [Revised: 04/18/2023] [Accepted: 04/23/2023] [Indexed: 05/01/2023]
Abstract
Amorphous solid dispersions (ASD) are one of the most adopted technologies for improving the solubility of novel molecules. Formulation of ASDs using solvent free methods such as hot melt extrusion (HME) has been in the spotlight off-lately. However, early-stage formulation development is tricky and a difficult bridge to pass due to limited drug availability. Material-sparing techniques (theoretical & practical) have been used for selecting suitable polymeric carriers for formulating ASDs. However, these techniques have limitations in predicting the effect of process parameters. The objective of this study is to use both theoretical and practical material-sparing techniques to optimize a polymer for the developing Triclabendazole (TBZ) ASDs. Initial screening by theoretical approaches suggested that TBZ is highly miscible with Kollidon®VA64 (VA64) and poorly miscible with Parteck®MXP (PVA). However, results from ASDs prepared using SCFe were opposite to these predictions. ASDs prepared using either technique and both VA64 and PVA showed >200x increase in solubility. Each formulation released >85% of drug in less than 15 mins. Although the thermodynamic phase diagram suggested that VA64 was the ideal polymer for TBZ-ASDs, it has certain limitations in factoring the different elements during melt-processing and hence, practical approaches like SCFe could help in predicting the drug-polymer miscibility for HME processing.
Collapse
Affiliation(s)
- Siddhant Palekar
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Hemanth K Mamidi
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA; Continuus Pharmaceuticals Inc, Woburn, MA, USA
| | - Yi Guo
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Richa Vartak
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Ketan Patel
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA.
| |
Collapse
|
3
|
Shukla A, Dumpa NR, Thakkar R, Shettar A, Ashour E, Bandari S, Repka MA. Influence of Poloxamer on the Dissolution and Stability of Hot-Melt Extrusion-Based Amorphous Solid Dispersions Using Design of Experiments. AAPS PharmSciTech 2023; 24:107. [PMID: 37100926 DOI: 10.1208/s12249-023-02562-3] [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: 02/03/2023] [Accepted: 04/02/2023] [Indexed: 04/28/2023] Open
Abstract
The current study aimed to see the effects of poloxamer P407 on the dissolution performance of hydroxypropyl methylcellulose acetate succinate (AquaSolve™ HPMC-AS HG)-based amorphous solid dispersions (ASD). A weakly acidic, poorly water-soluble active pharmaceutical ingredient (API), mefenamic acid (MA), was selected as a model drug. Thermal investigations, including thermogravimetry (TG) and differential scanning calorimetry (DSC), were conducted for raw materials and physical mixtures as a part of the pre-formulation studies and later to characterize the extruded filaments. The API was blended with the polymers using a twin shell V-blender for 10 min and then extruded using an 11-mm twin-screw co-rotating extruder. Scanning electron microscopy (SEM) was used to study the morphology of the extruded filaments. Furthermore, Fourier-transform infrared spectroscopy (FT-IR) was performed to check the intermolecular interactions of the components. Finally, to assess the in vitro drug release of the ASDs, dissolution testing was conducted in phosphate buffer (0.1 M, pH 7.4) and hydrochloric acid-potassium chloride (HCl-KCl) buffer (0.1 M, pH 1.2). The DSC studies confirmed the formation of the ASDs, and the drug content of the extruded filaments was observed to be within an acceptable range. Furthermore, the study concluded that the formulations containing poloxamer P407 exhibited a significant increase in dissolution performance compared to the filaments with only HPMC-AS HG (at pH 7.4). In addition, the optimized formulation, F3, was stable for over 3 months when exposed to accelerated stability studies.
Collapse
Affiliation(s)
- Ashay Shukla
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, 38677, Mississippi, USA
| | - Nagi Reddy Dumpa
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, 38677, Mississippi, USA
| | - Rishi Thakkar
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, 38677, Mississippi, USA
| | - Abhishek Shettar
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, 38677, Mississippi, USA
| | - Eman Ashour
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, 38677, Mississippi, USA
| | - Suresh Bandari
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, 38677, Mississippi, USA
| | - Michael A Repka
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, 38677, Mississippi, USA.
- Pii Center for Pharmaceutical Technology, The University of Mississippi, University, 38677, Mississippi, USA.
| |
Collapse
|
4
|
Andrés Real D, Gagliano A, Sonsini N, Wicky G, Orzan L, Leonardi D, Salomon C. Design and optimization of pH-sensitive Eudragit nanoparticles for improved oral delivery of triclabendazole. Int J Pharm 2022; 617:121594. [PMID: 35182705 DOI: 10.1016/j.ijpharm.2022.121594] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/10/2022] [Accepted: 02/12/2022] [Indexed: 11/28/2022]
Abstract
Design of Experiments (DoE) techniques were used to identify and optimize the parameters involved in the formulation of triclabendazole pH-sensitive Eudragit® nanoparticles (NPs). Using a Placket Burmann design, Eudragit® E, Eudragit® RS, and two stabilizers (PVP and PVA) were evaluated for NPs formulation by nanoprecipitation. Based on the screening results, Eudragit E 100® and PVP were selected as excipients, and their levels were studied and optimized using a central composite design, obtaining an optimum nanoparticulated system with a Size of 240 nm, a PDI of 0.420, and a ZP of 46.3 mV. Finally, a full characterization of the optimum system was carried out by XRD, DSC, equilibrium solubility, and dissolution rate in biorelevant mediums. As observed in XRD and DSC, the nanoencapsulation process produced a remarkable reduction in drug crystallinity that improved drug solubility and dissolution rate. Although more than 90% of TCBZ was dissolved in acidic mediums at 10 minutes, no increase in solubility or dissolution rate was observed in simulated saliva. Consequently, the development of pH-sensitive Eudragit® NPs would be a promising strategy in developing an immediate gastric release TCBZ formulation for oral delivery.
Collapse
Affiliation(s)
- Daniel Andrés Real
- Departamento de Química Farmacológica y Toxicológica, Universidad de Chile, Santos Dumont 964, 8380494 Santiago, Chile; Advanced Center of Chronic Diseases (ACCDiS), Universidad de Chile. Santos Dumont 964, Independencia, Santiago 8380494, Chile; Departamento de Farmacia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000, Rosario, Argentina
| | - Ailen Gagliano
- Departamento de Farmacia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000, Rosario, Argentina
| | - Nahuel Sonsini
- Departamento de Farmacia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000, Rosario, Argentina
| | - Gaspar Wicky
- Departamento de Farmacia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000, Rosario, Argentina
| | - Lucas Orzan
- Departamento de Farmacia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000, Rosario, Argentina
| | - Darío Leonardi
- Departamento de Farmacia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000, Rosario, Argentina; Instituto de Química de Rosario, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Suipacha 570, 2000, Rosario, Argentina
| | - Claudio Salomon
- Departamento de Farmacia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, 2000, Rosario, Argentina; Instituto de Química de Rosario, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Suipacha 570, 2000, Rosario, Argentina
| |
Collapse
|
5
|
Lopez-Vidal L, Real JP, Real DA, Camacho N, Kogan MJ, Paredes AJ, Palma SD. Nanocrystal-based 3D-printed tablets: Semi-solid extrusion using melting solidification printing process (MESO-PP) for oral administration of poorly soluble drugs. Int J Pharm 2022; 611:121311. [PMID: 34813905 DOI: 10.1016/j.ijpharm.2021.121311] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 01/01/2023]
Abstract
This is the first report on the inclusion of nanocrystals (NCs) within 3D-printed oral solid dosage forms -3D-printed tablets or printlets- produced by the Melting Solidification Printing Process (MESO-PP) 3D printing technique. This method allowed the incorporation of albendazole (ABZ) nanocrystals in a concentration of up to 50% w/w, something not achieved in conventional tablets. An ink of PEG 1500/propylenegycol was used as a carrier and no physicochemical interactions or crystallinity modifications were observed due to the inclusion of ABZ-NCs into the ink, as demonstrated by TGA, DSC, XRD and FT-IR. In particular, the relative crystallinity of the ink loaded with NCs was 97.8% similar to the physical mixture of the components. Moreover, the presence of NCs was observed in the surface and matrix of the printlets by SEM. In addition, the printlet NCs demonstrated to be more effective than NCs included in hard gelatin capsules in improving drug dissolution in HCl 0.1 N. The particle size, crystallinity and chemical stability of the nanocrystals was maintained before and after 180 days of storage. Thus, these findings exhibit relevant pharmaceutical potential for developing stable, fast-release, oral, solid dosage forms of poorly soluble drugs combining 3D printing and nanocrystals. Additionally, this technique could be applied for printing objects using different types of nanocrystals embedded in low melting temperature polymers.
Collapse
Affiliation(s)
- Lucía Lopez-Vidal
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), CONICET, Córdoba, Argentina; Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, X5000XHUA Córdoba, Argentina
| | - Juan Pablo Real
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), CONICET, Córdoba, Argentina; Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, X5000XHUA Córdoba, Argentina
| | - Daniel Andrés Real
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santos Dumont 964, Independencia, Santiago 8380494, Chile; Advanced Center for Chronic Diseases, ACCDiS, Santiago, Chile
| | - Nahuel Camacho
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), CONICET, Córdoba, Argentina; Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, X5000XHUA Córdoba, Argentina
| | - Marcelo J Kogan
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santos Dumont 964, Independencia, Santiago 8380494, Chile; Advanced Center for Chronic Diseases, ACCDiS, Santiago, Chile
| | - Alejandro J Paredes
- School of Pharmacy, Medical Biology Centre, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Santiago Daniel Palma
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), CONICET, Córdoba, Argentina; Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, X5000XHUA Córdoba, Argentina.
| |
Collapse
|
6
|
Alavi SE, Ebrahimi Shahmabadi H. Anthelmintics for drug repurposing: Opportunities and challenges. Saudi Pharm J 2021; 29:434-445. [PMID: 34135669 PMCID: PMC8180459 DOI: 10.1016/j.jsps.2021.04.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/03/2021] [Indexed: 12/14/2022] Open
Abstract
Drug repositioning is defined as a process to identify a new application for drugs. This approach is critical as it takes advantage of well-known pharmacokinetics, pharmacodynamics, and toxicity profiles of the drugs; thus, the chance of their future failure decreases, and the cost of their development and the required time for their approval are reduced. Anthelmintics, which are antiparasitic drugs, have recently demonstrated promising anticancer effects in vitro and in vivo. This literature review focuses on the potential of anthelmintics for repositioning in the treatment of cancers. It also discusses their pharmacokinetics and pharmacodynamics as antiparasitic drugs, proposed anticancer mechanisms, present development conditions, challenges in cancer therapy, and strategies to overcome these challenges.
Collapse
Affiliation(s)
- Seyed Ebrahim Alavi
- Department of Microbiology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hasan Ebrahimi Shahmabadi
- Department of Microbiology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
7
|
Khanfar M, Al-Remawi M, Al-Akayleh F, Hmouze S. Preparation and Evaluation of Co-amorphous Formulations of Telmisartan-Amino Acids as a Potential Method for Solubility and Dissolution Enhancement. AAPS PharmSciTech 2021; 22:112. [PMID: 33748914 DOI: 10.1208/s12249-021-01952-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 02/03/2021] [Indexed: 11/30/2022] Open
Abstract
Telmisartan (TLM) is a potent antihypertensive drug with pH-dependent aqueous solubility. This work aimed to enhance the solubility and dissolution rate of TLM by the co-amorphous drug amino acid (AA) approach by combining TLM, with different types and ratios of AAs. The co-amorphous TLM-AA blends were prepared by freeze-drying and investigated for solid-state characteristics like the dissolution rate enhancement of TLM. Among the prepared co-amorphous formulations, TLM-arginine (ARG) exhibited the greatest enhancement in solubility with increasing the molar ratio of ARG. The TLM-ARG at 1:2 ratio showed about a 57-fold increase in solubility of TLM and the highest dissolution percentage in phosphate buffer (pH7.5) (100% in 20 minutes) compared to both crystalline TLM (20% in 60 min) and physical mixture. Powder XRD, DSC, FTIR analysis and SEM demonstrated the formation of amorphous form within the co-amorphous formulations. Only TLM:ARG (1:0.5) were stable at (40°C, 75% RH) for a minimum of 90 days. In conclusion, ARG was able to stabilize the amorphous form of TLM and enhances its aqueous solubility and dissolution. The 1:2 w/w ratio of TLM-ARG co-amorphous showed the best solubility and dissolution rate while the 1:0.5 w/w ratio showed the best stability.
Collapse
|
8
|
A quality by design approach for optimization of Lecithin/Span® 80 based nanoemulsions loaded with hydrophobic drugs. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2020.114743] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
9
|
Bertoni S, Albertini B, Passerini N. Different BCS Class II Drug-Gelucire Solid Dispersions Prepared by Spray Congealing: Evaluation of Solid State Properties and In Vitro Performances. Pharmaceutics 2020; 12:pharmaceutics12060548. [PMID: 32545643 PMCID: PMC7356387 DOI: 10.3390/pharmaceutics12060548] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/05/2020] [Accepted: 06/10/2020] [Indexed: 12/17/2022] Open
Abstract
Delivery of poorly water soluble active pharmaceutical ingredients (APIs) by semi-crystalline solid dispersions prepared by spray congealing in form of microparticles (MPs) is an emerging method to increase their oral bioavailability. In this study, solid dispersions based on hydrophilic Gelucires® (Gelucire® 50/13 and Gelucire® 48/16 in different ratio) of three BCS class II model compounds (carbamazepine, CBZ, tolbutamide, TBM, and cinnarizine, CIN) having different physicochemical properties (logP, pKa, Tm) were produced by spray congealing process. The obtained MPs were investigated in terms of morphology, particles size, drug content, solid state properties, drug-carrier interactions, solubility, and dissolution performances. The solid-state characterization showed that the properties of the incorporated drug had a profound influence on the structure of the obtained solid dispersion: CBZ recrystallized in a different polymorphic form, TBM crystallinity was significantly reduced as a result of specific interactions with the carrier, while smaller crystals were observed in case of CIN. The in vitro tests suggested that the drug solubility was mainly influenced by carrier composition, while the drug dissolution behavior was affected by the API solid state in the MPs after the spray congealing process. Among the tested APIs, TBM-Gelucire dispersions showed the highest enhancement in drug dissolution as a result of the reduced drug crystallinity.
Collapse
|
10
|
Cheng X, Gao J, Li J, Cheng G, Zou M, Piao H. In Vitro-In Vivo Correlation for Solid Dispersion of a Poorly Water-Soluble Drug Efonidipine Hydrochloride. AAPS PharmSciTech 2020; 21:160. [PMID: 32476084 DOI: 10.1208/s12249-020-01685-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 04/11/2020] [Indexed: 12/11/2022] Open
Abstract
The aim of this present study was to investigate the ability of different dissolution methods to predict the in vivo performance of efonidipine hydrochloride (EFH). The solid dispersions of EFH were prepared by solvent evaporation method with HPMC-AS as matrix and urea as a pH adjusting agent. The paddle method, the open-loop, and the closed-loop flow-through cell methods were studied. In the study, Weibull's model was the best fit to explain release profiles. The pharmacokinetics behaviors of two kinds of solid dispersions with different release rate were investigated in comparison to the EFH after oral administration in rats. In vivo absorption was calculated by a numerical deconvolution method. In the study, the level A in vivo and in vitro correlation (IVIVC) was utilized. The correlation coefficient was calculated and interpreted by means of linear regression analysis (Origin.Pro.8.5 software). As a result, excellent IVIVC for solid dispersions and crude drug (r2 = 0.9352-0.9916) was obtained for the dissolution rate determined with flow-through cell open-loop system in phosphate buffer solution with 0.1% (w/v) polysorbate 80 at pH 6.5, the flow-rate of 4 mL/min. In addition, the self-assembled flow cell system had good repeatability and accuracy. The dissolution rate of the solid dispersion could be slowed down by the flow-through method, and the difference caused by preparation was significantly distinguished. The study demonstrated that flow-through cell method of the open-loop, compared with paddle method, was suitable for predicting in vivo performance of EFH solid dispersions.
Collapse
|
11
|
Aly UF, Sarhan HAM, Ali TFS, Sharkawy HAEB. Applying Different Techniques to Improve the Bioavailability of Candesartan Cilexetil Antihypertensive Drug. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1851-1865. [PMID: 32523332 PMCID: PMC7234962 DOI: 10.2147/dddt.s248511] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 04/27/2020] [Indexed: 01/31/2023]
Abstract
Purpose The objective of this study was to compare different techniques to enhance the solubility and dissolution rate, and hence the bioavailability of candesartan cilexetil. Methods To achieve this target, various techniques were employed such as solid dispersions, inclusion complexes, and preparation of candesartan nanoparticles. Following the preparations, all samples were characterized for their physicochemical properties, and the samples of the best results were subjected to further bioavailability studies. Results Results of dissolution studies revealed an increase in the dissolution rate of all samples. The highest dissolution rate was achieved using solid dispersion of the drug with PVP K-90 (1:4). Physicochemical investigations (XR, DSC, and FT-IR) suggested formation of hydrogen bonding and changing in the crystalline structure of the drug. Regarding the inclusion complexes, more stable complex was formed between HP-β-CD and CC compared to β-CD, as indicated by phase solubility diagrams. Antisolvent method resulted in the preparation of stable nanoparticles, as indicated by ζ potential, with average particle size of 238.9 ± 19.25 nm using PVP K-90 as a hydrophilic polymer. The best sample that gave the highest dissolution rate (CC/PVP K-90 1:4) was allowed for further pharmacokinetic studies using UPLC MS/MS assay of rabbit plasma. Results showed a significant increase in the bioavailability of CC from ~15% to ~48%. Conclusion The bioavailability of CC was significantly improved from ~15% to ~48% when formulated as SDs with PVP K-90 with 1:4 drug:polymer ratio.
Collapse
Affiliation(s)
- Usama Farghaly Aly
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, Egypt
| | | | - Taha F S Ali
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia, Egypt
| | | |
Collapse
|