1
|
Kapoor DU, Singh S, Sharma P, Prajapati BG. Amorphization of Low Soluble Drug with Amino Acids to Improve Its Therapeutic Efficacy: a State-of-Art-Review. AAPS PharmSciTech 2023; 24:253. [PMID: 38062314 DOI: 10.1208/s12249-023-02709-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 11/19/2023] [Indexed: 12/18/2023] Open
Abstract
Low aqueous solubility of drug candidates is an ongoing challenge and pharmaceutical manufacturers pay close attention to amorphization (AMORP) technology to improve the solubility of drugs that dissolve poorly. Amorphous drug typically exhibits much higher apparent solubility than their crystalline form due to high energy state that enable them to produce a supersaturated state in the gastrointestinal tract and thereby improve bioavailability. The stability and augmented solubility in co-amorphous (COA) formulations is influenced by molecular interactions. COA are excellent carriers-based drug delivery systems for biopharmaceutical classification system (BCS) class II and class IV drugs. The three important critical quality attributes, such as co-formability, physical stability, and dissolution performance, are necessary to illustrate the COA systems. New amorphous-stabilized carriers-based fabrication techniques that improve drug loading and degree of AMORP have been the focus of emerging AMORP technology. Numerous low-molecular-weight compounds, particularly amino acids such as glutamic acid, arginine, isoleucine, leucine, valine, alanine, glycine, etc., have been employed as potential co-formers. The review focus on the prevailing drug AMORP strategies used in pharmaceutical research, including in situ AMORP, COA systems, and mesoporous particle-based methods. Moreover, brief characterization techniques and the application of the different amino acids in stabilization and solubility improvements have been related.
Collapse
Affiliation(s)
| | - Sudarshan Singh
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Office of Research Administration, Faculty of Pharmacy, Chiang Mai University, 50200, Chiang Mai, Thailand.
| | - Pratishtha Sharma
- School of Pharmacy, Raffles University, Neemrana, Rajasthan, 301020, India
| | - Bhupendra G Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva, 384012, India.
| |
Collapse
|
2
|
Budiman A, Handini AL, Muslimah MN, Nurani NV, Laelasari E, Kurniawansyah IS, Aulifa DL. Amorphous Solid Dispersion as Drug Delivery Vehicles in Cancer. Polymers (Basel) 2023; 15:3380. [PMID: 37631436 PMCID: PMC10457821 DOI: 10.3390/polym15163380] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Cancer treatment has improved over the past decades, but a major challenge lies in drug formulation, specifically for oral administration. Most anticancer drugs have poor water solubility which can affect their bioavailability. This causes suboptimal pharmacokinetic performance, resulting in limited efficacy and safety when administered orally. As a result, it is essential to develop a strategy to modify the solubility of anticancer drugs in oral formulations to improve their efficacy and safety. A promising approach that can be implemented is amorphous solid dispersion (ASD) which can enhance the aqueous solubility and bioavailability of poorly water-soluble drugs. The addition of a polymer can cause stability in the formulations and maintain a high supersaturation in bulk medium. Therefore, this study aimed to summarize and elucidate the mechanisms and impact of an amorphous solid dispersion system on cancer therapy. To gather relevant information, a comprehensive search was conducted using keywords such as "anticancer drug" and "amorphous solid dispersion" in the PubMed, Scopus, and Google Scholar databases. The review provides an overview and discussion of the issues related to the ASD system used to improve the bioavailability of anticancer drugs based on molecular pharmaceutics. A thorough understanding of anticancer drugs in this system at a molecular level is imperative for the rational design of the products.
Collapse
Affiliation(s)
- Arif Budiman
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Annisa Luthfiyah Handini
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Mutia Nur Muslimah
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Neng Vera Nurani
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Eli Laelasari
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Insan Sunan Kurniawansyah
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Diah Lia Aulifa
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia;
| |
Collapse
|
3
|
Kestur U, Patel A, Badawy S, Mathias N, Zhang L. Strategies for Managing Solid Form Transformation Risk in Drug Product. J Pharm Sci 2023; 112:909-921. [PMID: 36513146 DOI: 10.1016/j.xphs.2022.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/03/2022] [Accepted: 12/04/2022] [Indexed: 12/14/2022]
Abstract
The International Conference of Harmonization (ICH) Q6A document provides guidance on setting specifications for new drug substances and drug products.1 In this paper we focus on decision trees 4 (#1) to (#3) in the guidance related to solid-state form transformation. Form transformation could occur from use of high energy forms to overcome solubility challenges or stresses from manufacturing processes. The decision trees provide guidance on when and how polymorphic form changes should be monitored and controlled. However, guidance is high level and does not capture aspects related to assessments needed to understand if there is a risk of transformation or tools that can be integrated to understand the severity of bioavailability impact at different stages of development. The objective of this paper is therefore to provide comprehensive chemistry manufacturing and controls (CMC) and regulatory strategies to manage the risk of form transformation. This includes practical workflows for form transformation risk assessment, analytical tools to detect and quantify the transformation including their shortcomings, biopharmaceutical tools to understand the severity of transformation risk and if needed justify the limits based on clinical relevance. Finally, a few case studies are discussed that capture how the workflow can be used to manage transformation risk.
Collapse
Affiliation(s)
- Umesh Kestur
- Drug Product Development, Bristol Myers Squibb, One Squibb Drive, New Brunswick, NJ 08903, USA.
| | - Anisha Patel
- Drug Product Development, Bristol Myers Squibb, One Squibb Drive, New Brunswick, NJ 08903, USA
| | - Sherif Badawy
- Drug Product Development, Bristol Myers Squibb, One Squibb Drive, New Brunswick, NJ 08903, USA
| | - Neil Mathias
- Drug Product Development, Bristol Myers Squibb, One Squibb Drive, New Brunswick, NJ 08903, USA
| | - Limin Zhang
- Analytical Strategy & Operations, Bristol Myers Squibb, One Squibb Drive, New Brunswick, NJ 08903, USA
| |
Collapse
|
4
|
Palatable Levocetirizine Dihydrochloride Solid Dispersed Fast-Dissolving Films: Formulation and In Vitro and In Vivo Characterization. ScientificWorldJournal 2022; 2022:1552602. [PMID: 36479553 PMCID: PMC9722282 DOI: 10.1155/2022/1552602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/11/2022] [Accepted: 09/14/2022] [Indexed: 11/30/2022] Open
Abstract
One of the most important issues for bitter-tasting drugs such as levocetirizine dihydrochloride (LCD) is the production of palatable dosage forms. LCD also has a delayed onset of action following oral administration. In this study, solid dispersed fast-dissolving films (FDFs) of LCD using the solvent casting method for oral application were prepared and evaluated. The FDF is composed of HPMC as the film forming polymer and different types of superdisintegrants (sodium starch glycolate, croscarmellose sodium, or crospovidone). FDF containing crospovidone showed the highest percentage release of the drug (100.54% ± 1.47 within 3 min.) and was chosen for fabricating into palatable solid dispersed FDFs using different ratios of gelatine. The results of Raman and FTIR revealed that the drug's crystalline structure has been disrupted, and the drug has intermolecular hydrogen bonds with gelatine. The solid dispersed FDF (LF-7), which contained the drug in the form of a 1 : 1 solid dispersion with gelatine, showed a rapid in vitro disintegration (25 seconds) and a burst release of the drug (99.22% ± 2.22 within one min). The in vivo studies were conducted on human participants and showed a significant (p < 0.05) reduction in disintegration time (9.43 ± 2.16 sec.) and higher taste masking ability of the solid dispersed FDF (LF-7) compared to the nonsolid dispersed FDF (LF-4). The stability studies indicated that the prepared FDF remained stable over three months. Overall, FDFs of levocetirizine dihydrochloride with a palatable and rapid onset of action were developed to relieve allergic symptoms.
Collapse
|
5
|
Bioavailability of Celecoxib Formulated with Mesoporous Magnesium Carbonate-An In Vivo Evaluation. Molecules 2022; 27:molecules27196188. [PMID: 36234733 PMCID: PMC9570901 DOI: 10.3390/molecules27196188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
An attractive approach to increase the aqueous apparent solubility of poorly soluble drugs is to formulate them in their amorphous state. In the present study, celecoxib, a poorly soluble drug, was successfully loaded into mesoporous magnesium carbonate (MMC) in its amorphous state via a solvent evaporation method. Crystallization of celecoxib was suppressed, and no reaction with the carrier was detected. The MMC formulation was evaluated in vitro and in vivo in terms of oral bioavailability. Celebra®, a commercially available formulation, was used as a reference. The two celecoxib formulations were orally administrated in male rats (average of n = 6 animals per group), and blood samples for plasma were taken from all animals at different time points after administration. There was no statistical difference (p > 0.05) in AUCinf between the two formulations. The results showed that MMC may be a promising drug delivery excipient for increasing the bioavailability of compounds with solubility-limited absorption.
Collapse
|
6
|
Wolbert F, Nikoleit K, Steinbrink M, Luebbert C, Sadowski G. The Shelf Life of ASDs: 1. Measuring the Crystallization Kinetics at Humid Conditions. Mol Pharm 2022; 19:2483-2494. [PMID: 35729680 DOI: 10.1021/acs.molpharmaceut.2c00188] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Amorphous solid dispersions (ASDs), where an active pharmaceutical ingredient (API) is dissolved in a polymer, are a favored formulation technique to achieve sufficient bioavailability of poorly water-soluble APIs. The shelf life of such ASDs is often limited by API crystallization. Crystallization depends strongly on the storage conditions (relative humidity and temperature) and the polymer selected for generating the ASD. Determining the crystallization kinetics of ASDs under various conditions requires suitable analytical methods. In this work, two different analytical methods were compared and cross-validated: The first builds on water-sorption measurements combined with thermodynamic predictions ( Eur. J. Pharm. Biopharm. 2018, 127, 183-193, DOI: 10.1016/j.toxrep.2018.11.002), whereas the second applies Raman spectroscopy. Using the two independent methods, factors influencing the crystallization kinetics of ASDs containing the API griseofulvin were investigated quantitatively. It was found that crystallization kinetics increases with increasing temperature and relative humidity. Additionally, the influence of different polymers (poly(vinylpyrrolidone-co-vinyl acetate) and Soluplus) on crystallization kinetics were investigated. The experimentally obtained crystallization kinetics were described using the Johnson-Mehl-Avrami-Kolmogorov model and are the basis for future shelf life predictions at desired storage conditions.
Collapse
Affiliation(s)
- Friederike Wolbert
- INVITE GmbH, Drug Delivery Innovation Center (DDiC), 51368 Leverkusen, Germany.,TU Dortmund University, Department of Biochemical and Chemical Engineering, Laboratory of Thermodynamics, Emil-Figge-Strasse 70, D-44227 Dortmund, Germany
| | - Klaudia Nikoleit
- TU Dortmund University, Department of Biochemical and Chemical Engineering, Laboratory of Thermodynamics, Emil-Figge-Strasse 70, D-44227 Dortmund, Germany
| | - Miklas Steinbrink
- TU Dortmund University, Department of Biochemical and Chemical Engineering, Laboratory of Thermodynamics, Emil-Figge-Strasse 70, D-44227 Dortmund, Germany
| | - Christian Luebbert
- TU Dortmund University, Department of Biochemical and Chemical Engineering, Laboratory of Thermodynamics, Emil-Figge-Strasse 70, D-44227 Dortmund, Germany
| | - Gabriele Sadowski
- TU Dortmund University, Department of Biochemical and Chemical Engineering, Laboratory of Thermodynamics, Emil-Figge-Strasse 70, D-44227 Dortmund, Germany
| |
Collapse
|
7
|
Gabrič A, Hodnik Ž, Pajk S. Oxidation of Drugs during Drug Product Development: Problems and Solutions. Pharmaceutics 2022; 14:pharmaceutics14020325. [PMID: 35214057 PMCID: PMC8876153 DOI: 10.3390/pharmaceutics14020325] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 11/16/2022] Open
Abstract
Oxidation is the second most common degradation pathway for pharmaceuticals, after hydrolysis. However, in contrast to hydrolysis, oxidation is mechanistically more complex and produces a wider range of degradation products; oxidation is thus harder to control. The propensity of a drug towards oxidation is established during forced degradation studies. However, a more realistic insight into degradation in the solid state can be achieved with accelerated studies of mixtures of drugs and excipients, as the excipients are the most common sources of impurities that have the potential to initiate oxidation of a solid drug product. Based on the results of these studies, critical parameters can be identified and appropriate measures can be taken to avoid the problems that oxidation poses to the quality of a drug product. This article reviews the most common types of oxidation mechanisms, possible sources of reactive oxygen species, and how to minimize the oxidation of a solid drug product based on a well-planned accelerated study.
Collapse
Affiliation(s)
- Alen Gabrič
- Krka d.d., R&D, Šmarješka Cesta 6, 8001 Novo Mesto, Slovenia;
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000 Ljubljana, Slovenia
| | - Žiga Hodnik
- Krka d.d., R&D, Šmarješka Cesta 6, 8001 Novo Mesto, Slovenia;
- Correspondence: (Ž.H.); (S.P.)
| | - Stane Pajk
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000 Ljubljana, Slovenia
- Correspondence: (Ž.H.); (S.P.)
| |
Collapse
|
8
|
Increasing the Transport of Celecoxib over a Simulated Intestine Cell Membrane Model Using Mesoporous Magnesium Carbonate. Molecules 2021; 26:molecules26216353. [PMID: 34770762 PMCID: PMC8588146 DOI: 10.3390/molecules26216353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/09/2021] [Accepted: 10/18/2021] [Indexed: 12/03/2022] Open
Abstract
In the current work, mesoporous magnesium carbonate (MMC) was used to suppress crystallization of the poorly soluble drug celecoxib (CXB). This resulted in both a higher dissolution rate and supersaturation of the substance in vitro as well as an increased transfer of CXB over a Caco-2 cell membrane mimicking the membrane in the small intestine. The CXB flux over the cell membrane showed a linear behavior over the explored time period. These results indicate that MMC may be helpful in increasing the bioavailability and obtaining a continuous release of CXB, and similar substances, in vivo. Neusilin US2 was used as a reference material and showed a more rapid initial release with subsequent crystallization of the incorporated CXB in the release media. The presented results form the foundation of future development of MMC as a potential carrier for poorly soluble drugs.
Collapse
|
9
|
Lapuk SE, Mukhametzyanov TA, Schick C, Gerasimov AV. Crystallization kinetics and glass-forming ability of rapidly crystallizing drugs studied by Fast Scanning Calorimetry. Int J Pharm 2021; 599:120427. [PMID: 33662469 DOI: 10.1016/j.ijpharm.2021.120427] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/09/2021] [Accepted: 02/20/2021] [Indexed: 11/16/2022]
Abstract
The use of the amorphous forms of drugs is a modern approach for the enhancement of bioavailability. At the same time, the high cooling rate needed to obtain the metastable amorphous state often prevents its investigation using conventional laboratory methods such as differential scanning calorimetry, X-ray powder diffractometry. One of the ways to overcome this problem may be the application of Fast Scanning Calorimetry. This method allows direct determination of the critical cooling rate of the melt and kinetic parameters of the crystallization for bad glass formers. In the present work, the amorphous states of dopamine hydrochloride and atenolol were created using Fast Scanning Calorimetry for the first time. Critical cooling rates and glass transition temperatures of these drugs were determined. Based on the values of the kinetic fragility parameter, dopamine hydrochloride glass can be considered strong, while atenolol glass is moderately strong. Both model-based and model-free approaches were employed to determine the kinetic parameters of cold crystallization of dopamine and atenolol. The results were compared with the data from isothermal crystallization experiments. The Nakamura crystallization model provides the best description of the crystallization process and can be used to predict the long term stability of the amorphous forms of the drugs. The presented approaches may find applications in predicting the storage time and choosing the optimal storage conditions of the amorphous drugs prone to crystallization.
Collapse
Affiliation(s)
- S E Lapuk
- Department of Physical Chemistry, A.M. Butlerov Institute of Chemistry, Kazan Federal University, 420008, Kremlevskaya, 18, Kazan, Russia
| | - T A Mukhametzyanov
- Department of Physical Chemistry, A.M. Butlerov Institute of Chemistry, Kazan Federal University, 420008, Kremlevskaya, 18, Kazan, Russia
| | - C Schick
- Department of Physical Chemistry, A.M. Butlerov Institute of Chemistry, Kazan Federal University, 420008, Kremlevskaya, 18, Kazan, Russia
| | - A V Gerasimov
- Department of Physical Chemistry, A.M. Butlerov Institute of Chemistry, Kazan Federal University, 420008, Kremlevskaya, 18, Kazan, Russia.
| |
Collapse
|
10
|
Griffin SR, Takanti N, Sarkar S, Song Z, Vogt AD, Danzer GD, Simpson GJ. Disparities of Single-Particle Growth Rates in Buried Versus Exposed Ritonavir Crystals within Amorphous Solid Dispersions. Mol Pharm 2020; 17:4564-4571. [PMID: 33151697 DOI: 10.1021/acs.molpharmaceut.0c00744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Seeded growth rates of ritonavir in copovidone at 75% relative humidity (RH) and 50 °C were evaluated by single-particle tracking second harmonic generation (SHG) microscopy and found to be ∼3-fold slower for crystallites at the surface compared to the bulk. The shelf lives of final dosage forms containing amorphous solid dispersions (ASDs) are often dictated by the rates of active pharmaceutical ingredient crystallization. Upon exposure to elevated RH, the higher anticipated water content near the surfaces of ASDs has the potential to substantially impact nucleation and growth kinetics relative to the bulk. However, quantitative assessment of these differences in growth rates is complicated by challenges associated with discrimination of the two contributions (supersaturation and molecular mobility) in ensemble-averaged measurements. In the present study, "sandwich" materials were prepared, in which sparse populations of ritonavir single-crystalline seeds were pressed between two similar ASD films to assess bulk crystallization rates. These sandwich materials were compared and contrasted with analogously prepared "open-faced" samples, without the capping film, to assess the surface crystallization rates. Single-particle analysis by SHG microscopy time-series during in situ crystallization produced average growth rates of 3.8 μm/h for bulk columnar crystals with a particle-to-particle standard deviation of 0.9 μm/h. In addition, columnar crystal growth rates for surface particles were measured to be 1.3 μm/h and radiating crystal growth rates for surface particles were measured to be 1.0 μm/h, both with a particle-to-particle deviation of 0.4 μm/h. The observed appearance of radiating crystals upon surface seeding is attributed to reduced ritonavir solubility upon water adsorption at the interface, leading to higher defect densities in crystal growth. Despite substantial differences in crystal habit, correction of the surface growth rates by a factor of 4 from geometric effects resulted in relatively minor but statistically significant differences in the growth kinetics for the two local environments. These results are consistent, with viscosity being a relatively weak function of water absorption coupled with primarily diffusion-limited growth kinetics.
Collapse
Affiliation(s)
- Scott R Griffin
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| | - Nita Takanti
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| | - Sreya Sarkar
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| | - Zhengtian Song
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| | - Andrew D Vogt
- AbbVie Inc., 1 N Waukegan Road, North Chicago, Illinois 60064, United States
| | - Gerald D Danzer
- AbbVie Inc., 1 N Waukegan Road, North Chicago, Illinois 60064, United States
| | - Garth J Simpson
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|
11
|
Lapuk S, Mukhametzyanov T, Schick C, Gerasimov A. Kinetic stability of amorphous dipyridamole: A fast scanning calorimetry investigation. Int J Pharm 2020; 574:118890. [DOI: 10.1016/j.ijpharm.2019.118890] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/15/2019] [Accepted: 11/16/2019] [Indexed: 01/10/2023]
|
12
|
Hurley D, Carter D, Foong Ng LY, Davis M, Walker GM, Lyons JG, Higginbotham CL. An investigation of the inter-molecular interaction, solid-state properties and dissolution properties of mixed copovidone hot-melt extruded solid dispersions. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101132] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
13
|
Pawar J, Suryawanshi D, Moravkar K, Aware R, Shetty V, Maniruzzaman M, Amin P. Study the influence of formulation process parameters on solubility and dissolution enhancement of efavirenz solid solutions prepared by hot-melt extrusion: a QbD methodology. Drug Deliv Transl Res 2019; 8:1644-1657. [PMID: 29426975 DOI: 10.1007/s13346-018-0481-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The current study investigates the dissolution rate performance of amorphous solid solutions of a poorly water-soluble drug, efavirenz (EFV), in amorphous Soluplus® (SOL) and Kollidon® VA 64 (KVA64) polymeric systems. For the purpose of the study, various formulations with varying drug loadings of 30, 50, and 70% w/w were developed via hot-melt extrusion processing and adopting a Box-Behnken design of experiment (DoE) approach. The polymers were selected based on the Hansen solubility parameter calculation and the prediction of the possible drug-polymer miscibility. In DoE experiments, a Box-Behnken factorial design was conducted to evaluate the effect of independent variables such as Soluplus® ratio (A1), HME screw speed (A2), and processing temperature (A3), and Kollidon®VA64 ratio (B1), screw speed (B2), and processing temperature (B3) on responses such as solubility (X1 and Y1) and dissolution rate (X2 and Y2) for both ASS [EFV:SOL] and BSS [EFV:KVA64] systems. DSC and XRD data confirmed that bulk crystalline EFV transformed to amorphous form during the HME processing. Advanced chemical analyses conducted via 2D COSY NMR, FTIR chemical imaging, AFM analysis, and FTIR showed that EFV was homogenously dispersed in the respective polymer matrices. The maximum solubility and dissolution rate was observed in formulations containing 30% EFV with both SOL and KVA64 alone. This could be attributed to the maximum drug-polymer miscibility in the optimized formulations. The actual and predicted values of both responses were found precise and close to each other.
Collapse
Affiliation(s)
- Jaywant Pawar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, University under Section-3 of UGC Act-1956, Elite Status & Centre of Excellence - Govt. of Maharashtra, N. P. Marg, Matunga, Mumbai, 400019, India.
| | - Dilipkumar Suryawanshi
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, University under Section-3 of UGC Act-1956, Elite Status & Centre of Excellence - Govt. of Maharashtra, N. P. Marg, Matunga, Mumbai, 400019, India
| | - Kailas Moravkar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, University under Section-3 of UGC Act-1956, Elite Status & Centre of Excellence - Govt. of Maharashtra, N. P. Marg, Matunga, Mumbai, 400019, India
| | - Rahul Aware
- ACG Pharma Technologies Pvt. Ltd., Shirwal, Pune, Maharashtra, India
| | - Vasant Shetty
- ACG Pharma Technologies Pvt. Ltd., Shirwal, Pune, Maharashtra, India
| | - Mohammed Maniruzzaman
- Department of Pharmacy/Chemistry, School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9QJ, UK
| | - Purnima Amin
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, University under Section-3 of UGC Act-1956, Elite Status & Centre of Excellence - Govt. of Maharashtra, N. P. Marg, Matunga, Mumbai, 400019, India
| |
Collapse
|
14
|
Luebbert C, Sadowski G. In-situ determination of crystallization kinetics in ASDs via water sorption experiments. Eur J Pharm Biopharm 2018; 127:183-193. [DOI: 10.1016/j.ejpb.2018.02.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/19/2018] [Accepted: 02/20/2018] [Indexed: 11/28/2022]
|
15
|
Song Z, Sarkar S, Vogt AD, Danzer GD, Smith CJ, Gualtieri EJ, Simpson GJ. Kinetic Modeling of Accelerated Stability Testing Enabled by Second Harmonic Generation Microscopy. Anal Chem 2018; 90:4406-4413. [DOI: 10.1021/acs.analchem.7b04260] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Zhengtian Song
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47906, United States
- AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Sreya Sarkar
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47906, United States
- AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Andrew D. Vogt
- AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Gerald D. Danzer
- AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Casey J. Smith
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47906, United States
| | - Ellen J. Gualtieri
- Formulatrix, Inc., 10 DeAngelo Drive, Bedford, Massachusetts 01730, United States
| | - Garth J. Simpson
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47906, United States
| |
Collapse
|
16
|
Hurley D, Potter CB, Walker GM, Higginbotham CL. Investigation of Ethylene Oxide-co-propylene Oxide for Dissolution Enhancement of Hot-Melt Extruded Solid Dispersions. J Pharm Sci 2018; 107:1372-1382. [PMID: 29410037 DOI: 10.1016/j.xphs.2018.01.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/11/2018] [Accepted: 01/12/2018] [Indexed: 11/18/2022]
Abstract
The optimal design of amorphous solid dispersion formulations requires the use of excipients to maintain supersaturation and improve physical stability to ensure shelf-life stability and better absorption during intestinal transit, respectively. Blends of excipients (surfactants and polymers) are often used within pharmaceutical products to improve the oral delivery of Biopharmaceutical Classification System class II drugs. Therefore, in this study, a dissolution enhancer, poloxamer 407 (P407), was investigated to determine its effect on the dissolution properties and on the amorphous nature of the active pharmaceutical ingredient contained in the formulation. Phase solubility studies of indomethacin (INM) in aqueous solutions of P407 and poly(vinylpyrrolidone-vinyl acetate copolymer) showed an increase in the kinetic solubility of INM compared with the pure drug at 37°C with a Ka value of 0.041 μg/mL. The solid dispersions showed a higher dissolution rate when compared to pure and amorphous drugs when performed in pH buffer 1.2 with a kinetic solubility of 21 μg/mL. The stability data showed that the amorphous drug in solid solutions with poly(vinylpyrrolidone-vinyl acetate copolymer) and P407 remained amorphous, and the %P407 loading had no effect on the amorphous stability of INM. This study concluded that the amorphous solid dispersion contributed to the increased solubility of INM.
Collapse
Affiliation(s)
- Dean Hurley
- Materials Research Institute, Athlone Institute of Technology, Westmeath, Ireland
| | - Catherine B Potter
- Synthesis and Solid State Pharmaceutical Centre (SSPC), Bernal Institute, University of Limerick, Limerick, Ireland
| | - Gavin M Walker
- Synthesis and Solid State Pharmaceutical Centre (SSPC), Bernal Institute, University of Limerick, Limerick, Ireland
| | | |
Collapse
|
17
|
Luebbert C, Wessner M, Sadowski G. Mutual Impact of Phase Separation/Crystallization and Water Sorption in Amorphous Solid Dispersions. Mol Pharm 2018; 15:669-678. [DOI: 10.1021/acs.molpharmaceut.7b01076] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Christian Luebbert
- Department of Biochemical
and Chemical Engineering, Laboratory of Thermodynamics, TU Dortmund University, Emil-Figge-Straße 70, D-44227 Dortmund, Germany
| | - Maximilian Wessner
- Department of Biochemical
and Chemical Engineering, Laboratory of Thermodynamics, TU Dortmund University, Emil-Figge-Straße 70, D-44227 Dortmund, Germany
| | - Gabriele Sadowski
- Department of Biochemical
and Chemical Engineering, Laboratory of Thermodynamics, TU Dortmund University, Emil-Figge-Straße 70, D-44227 Dortmund, Germany
| |
Collapse
|
18
|
Calvo NL, Maggio RM, Kaufman TS. Chemometrics-assisted solid-state characterization of pharmaceutically relevant materials. Polymorphic substances. J Pharm Biomed Anal 2017; 147:518-537. [PMID: 28668295 DOI: 10.1016/j.jpba.2017.06.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/07/2017] [Accepted: 06/12/2017] [Indexed: 11/26/2022]
Abstract
Current regulations command to properly characterize pharmaceutically relevant solid systems. Chemometrics comprise a range of valuable tools, suitable to process large amounts of data and extract valuable information hidden in their structure. This review aims to detail the results of the fruitful association between analytical techniques and chemometrics methods, focusing on those which help to gain insight into the characteristics of drug polymorphism as an important aspect of the solid state of bulk drugs and drug products. Hence, the combination of Raman, terahertz, mid- and near- infrared spectroscopies, as well as instrumental signals resulting from X-ray powder diffraction, 13C solid state nuclear magnetic resonance spectroscopy and thermal methods with quali-and quantitative chemometrics methodologies are examined. The main issues reviewed, concerning pharmaceutical drug polymorphism, include the use of chemometrics-based approaches to perform polymorph classification and assignment of polymorphic identity, as well as the determination of given polymorphs in simple mixtures and complex systems. Aspects such as the solvation/desolvation of solids, phase transformation, crystallinity and the recrystallization from the amorphous state are also discussed. A brief perspective of the field for the next future is provided, based on the developments of the last decade and the current state of the art of analytical instrumentation and chemometrics methodologies.
Collapse
Affiliation(s)
- Natalia L Calvo
- Instituto de Química Rosario (IQUIR, CONICET-UNR) and Área Análisis de Medicamentos, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, Rosario (S2002LRK), Argentina
| | - Rubén M Maggio
- Instituto de Química Rosario (IQUIR, CONICET-UNR) and Área Análisis de Medicamentos, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, Rosario (S2002LRK), Argentina
| | - Teodoro S Kaufman
- Instituto de Química Rosario (IQUIR, CONICET-UNR) and Área Análisis de Medicamentos, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531, Rosario (S2002LRK), Argentina.
| |
Collapse
|
19
|
Kinoshita R, Ohta T, Shiraki K, Higashi K, Moribe K. Effects of wet-granulation process parameters on the dissolution and physical stability of a solid dispersion. Int J Pharm 2017; 524:304-311. [DOI: 10.1016/j.ijpharm.2017.04.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/22/2017] [Accepted: 04/04/2017] [Indexed: 11/26/2022]
|
20
|
Aucamp M, Milne M, Liebenberg W. Amorphous Sulfadoxine: A Physical Stability and Crystallization Kinetics Study. AAPS PharmSciTech 2016; 17:1100-9. [PMID: 26531745 DOI: 10.1208/s12249-015-0436-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/14/2015] [Indexed: 11/30/2022] Open
Abstract
Poor aqueous solubility of drugs and the improvement thereof has always been a challenge for the pharmaceutical industry. With this, one of the focuses of the pharmaceutical research scientist involves investigating possible metastable forms of a given drug to be incorporated into solid dosage forms. The rationale being, the improved solubility offered by the metastable solid-state forms of drugs. Solubility remains a major challenge for formulation scientists, especially with antimicrobial agents where the emergence of resistance is directly dependent on the concentration and duration of the parasite exposed to the drug. Sulfadoxine-pyrimethamine combination therapies are still the recommended treatments for uncomplicated Plasmodium falciparum malaria. The aim of this study was to prepare an amorphous form of sulfadoxine and to investigate the stability and recrystallization behavior thereof. The amorphous form was prepared by the well-known quench cooling of the melt. The physico-chemical properties and stability of amorphous sulfadoxine were studied using hot-stage microscopy (HSM), scanning electron microscopy (SEM), x-ray powder diffractometry (XRPD), differential scanning calorimetry (DSC), thermogravimetric analysis (TGA), as well as microcalorimetry. The recrystallization kinetics were studied isothermally by applying the Johnson-Mehl-Avrami model and non-isothermally by applying the Kissinger model. The physical stabilization of the amorphous form was investigated using physical mixtures of amorphous sulfadoxine with polyvinylpyrrolidone-25 (PVP-25). It was proved that sulfadoxine is a good glass former with relative high physical stability; however, water acts as a strong plasticizer for amorphous sulfadoxine, detrimentally affecting the stability during exposure to high moisture conditions.
Collapse
|
21
|
Zhang P, Zardán Gómez de la Torre T, Welch K, Bergström C, Strømme M. Supersaturation of poorly soluble drugs induced by mesoporous magnesium carbonate. Eur J Pharm Sci 2016; 93:468-74. [DOI: 10.1016/j.ejps.2016.08.059] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 08/25/2016] [Accepted: 08/29/2016] [Indexed: 11/15/2022]
|
22
|
Polymeric Amorphous Solid Dispersions: A Review of Amorphization, Crystallization, Stabilization, Solid-State Characterization, and Aqueous Solubilization of Biopharmaceutical Classification System Class II Drugs. J Pharm Sci 2016; 105:2527-2544. [DOI: 10.1016/j.xphs.2015.10.008] [Citation(s) in RCA: 557] [Impact Index Per Article: 69.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
23
|
Hédoux A. Recent developments in the Raman and infrared investigations of amorphous pharmaceuticals and protein formulations: A review. Adv Drug Deliv Rev 2016; 100:133-46. [PMID: 26686831 DOI: 10.1016/j.addr.2015.11.021] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 11/26/2015] [Accepted: 11/30/2015] [Indexed: 10/22/2022]
Abstract
The success rate for drug discovery and the development of innovative therapeutic strategies are intimately related to the physical properties of the solid-state condensed matter, which have direct influence on the bioavailability of Active Pharmaceutical Ingredients. In order to transform a new molecule in efficient drug, the material is brought into an amorphous state using various manufacturing processes including freeze drying, spray drying, hot melt extrusion and loading in different delivery devices. The infrared and Raman spectroscopic analyses used for exploring disordered and amorphous states, for the monitoring of the drug physical stability in drug delivery systems are described in this review.
Collapse
|
24
|
Zhang P, Zardán Gómez de la Torre T, Forsgren J, Bergström CA, Strømme M. Diffusion-Controlled Drug Release From the Mesoporous Magnesium Carbonate Upsalite ®. J Pharm Sci 2016; 105:657-663. [DOI: 10.1002/jps.24553] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/29/2015] [Accepted: 06/02/2015] [Indexed: 11/12/2022]
|
25
|
Ueda H, Muranushi N, Sakuma S, Ida Y, Endoh T, Kadota K, Tozuka Y. A Strategy for Co-former Selection to Design Stable Co-amorphous Formations Based on Physicochemical Properties of Non-steroidal Inflammatory Drugs. Pharm Res 2015; 33:1018-29. [PMID: 26700604 DOI: 10.1007/s11095-015-1848-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 12/17/2015] [Indexed: 12/30/2022]
Abstract
PURPOSE This study aimed to investigate the physicochemical factors contributing to stable co-amorphous formations and to design a co-former selection strategy. METHODS Non-steroidal inflammatory drugs were used as main components and/or co-formers. Physical mixtures of the materials were melted. Co-amorphization was characterized by the inhibition effect of the co-former on crystallization of the main component from the undercooled melt. The contribution of physicochemical factors to the co-amorphous formation was analyzed by multivariate analysis. Co-amorphous samples prepared by melting were subjected to thermal and spectroscopic analyses and the isothermal crystallization test. RESULTS Naproxen (NAP) was employed as the main component having a rapid crystallization tendency. Some materials used as the co-former inhibited the crystallization of amorphous NAP; decreasing melting temperatures of the components was an indicator of co-amorphization. The contribution of some physicochemical features (e.g., crystallization tendency, glass transition temperature (Tg)/melting temperature and molecular flexibility) of the co-formers to a co-amorphous formation was suggested by multivariate analysis. Deviation of the glass transition temperature from the theoretical value and changes in the infrared spectra of the co-amorphous samples were correlated with intermolecular interaction. The crystallization behaviors of the co-amorphous samples depended on their Tg. CONCLUSIONS The results showed a relationship between stable co-amorphous formation and the physicochemical features of the components, which should inform efficient co-former selection to design stable co-amorphous formations.
Collapse
Affiliation(s)
- Hiroshi Ueda
- Physicochemical and Preformulation, Applied Chemistry and Analysis, Research Laboratory for Development, Shionogi & Co., Ltd, 3-1-1, Futaba-cho, Toyonaka-shi, Osaka, 561-0825, Japan. .,Laboratory of Formulation Design and Pharmaceutical Technology, Osaka University of Pharmaceutical Sciences, 4-20-1, Nasahara, Takatsuki-shi, Osaka, 569-1094, Japan.
| | - Noriyuki Muranushi
- New Technology Department, Formulation Development Center, CMC Development Laboratories, Shionogi & Co., Ltd., 2-1-3, Kuise-Terajima, Amagasaki-shi, Hyogo, 660-0813, Japan
| | - Satoshi Sakuma
- New Technology Department, Formulation Development Center, CMC Development Laboratories, Shionogi & Co., Ltd., 2-1-3, Kuise-Terajima, Amagasaki-shi, Hyogo, 660-0813, Japan
| | - Yasuo Ida
- Physicochemical and Preformulation, Applied Chemistry and Analysis, Research Laboratory for Development, Shionogi & Co., Ltd, 3-1-1, Futaba-cho, Toyonaka-shi, Osaka, 561-0825, Japan
| | - Takeshi Endoh
- Physicochemical and Preformulation, Applied Chemistry and Analysis, Research Laboratory for Development, Shionogi & Co., Ltd, 3-1-1, Futaba-cho, Toyonaka-shi, Osaka, 561-0825, Japan
| | - Kazunori Kadota
- Laboratory of Formulation Design and Pharmaceutical Technology, Osaka University of Pharmaceutical Sciences, 4-20-1, Nasahara, Takatsuki-shi, Osaka, 569-1094, Japan
| | - Yuichi Tozuka
- Laboratory of Formulation Design and Pharmaceutical Technology, Osaka University of Pharmaceutical Sciences, 4-20-1, Nasahara, Takatsuki-shi, Osaka, 569-1094, Japan
| |
Collapse
|
26
|
Ali AA, Hashim AM. Evolution of Zinc Oxide Nanostructures Grown on Graphene by Ultrasonic Spray Pyrolysis and Its Statistical Growth Modelling. NANOSCALE RESEARCH LETTERS 2015; 10:452. [PMID: 26608535 PMCID: PMC4659785 DOI: 10.1186/s11671-015-1163-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 11/15/2015] [Indexed: 06/05/2023]
Abstract
The evolution of zinc oxide nanostructures grown on graphene by alcohol-assisted ultrasonic spray pyrolysis was investigated. The evolution of structures is strongly depended on pyrolysis parameters, i.e., precursor molarity, precursor flow rate, precursor injection/deposition time, and substrate temperature. Field-effect scanning electron microscope analysis, energy dispersive X-ray spectroscopy, X-ray diffraction, and transmission electron microscopy were used to investigate the properties of the synthesized nanostructures and to provide evidence for the structural changes according to the changes in the pyrolysis parameters. The optimum parameters to achieve maximum density and well-defined hexagonally shaped nanorods were a precursor molarity of 0.2 M, an injection flow rate of 6 ml/min, an injection time of 10 min, and a substrate temperature of 250-355 °C. Based on the experimental results, the response surface methodology (RSM) was used to model and optimize the independent pyrolysis parameters using the Box-Behnken design. Here, the responses, i.e., the nanostructure density, size, and shape factor, are evaluated. All of the computations were performed using the Design-Expert software package. Analysis of variance (ANOVA) was used to evaluate the results of the model and to determine the significant values for the independent pyrolysis parameters. The evolution of zinc oxide (ZnO) structures are well explained by the developed modelling which confirms that RSM is a reliable tool for the modelling and optimization of the pyrolysis parameters and prediction of nanostructure sizes and shapes.
Collapse
Affiliation(s)
- Amgad Ahmed Ali
- Malaysia-Japan International Institute of Technology, Universiti Teknologi Malaysia, Jalan Sultan Yahya Petra, 54100, Kuala Lumpur, Malaysia.
| | - Abdul Manaf Hashim
- Malaysia-Japan International Institute of Technology, Universiti Teknologi Malaysia, Jalan Sultan Yahya Petra, 54100, Kuala Lumpur, Malaysia.
| |
Collapse
|
27
|
Chen X, Stoneburner K, Ladika M, Kuo TC, Kalantar TH. High-Throughput Raman Spectroscopy Screening of Excipients for the Stabilization of Amorphous Drugs. APPLIED SPECTROSCOPY 2015; 69:1271-1280. [PMID: 26647050 DOI: 10.1366/15-07914] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Low aqueous solubility of active pharmaceutical ingredients (APIs) is an enduring problem in pharmaceutical development, and it is becoming increasingly prevalent among new drug candidates. It is estimated that about 40% of drugs in the development pipeline and approximately 60% of the drugs coming directly from discovery suffer from poor aqueous solubility and slow dissolution, thereby reducing their bioavailability and efficacy and thus preventing their commercialization. It is well known that utilizing the amorphous form of a drug can be a useful approach to improve the dissolution rate and solubility of poorly water-soluble APIs. Amorphous compounds are thermodynamically unstable, but they can be stabilized by combining them with a carrier polymer (excipient) to form a solid dispersion. High-throughput Raman spectroscopy was used in this study to identify excipients that promote formation and stabilization of the amorphous drug form in solid dispersions. Four model APIs were used as poorly soluble drug candidates: ketoprofen, danazol, griseofulvin, and probucol. The Raman signals of excipients were generally negligible, and therefore Raman bands from the drugs were used with minimal spectral pre-processing. By comparing Raman spectra collected from the APIs in the crystalline and molten state, appropriate spectral features and regions were identified for the development of semi-quantitative methods to determine the amorphous content for each API. It is demonstrated that methods based on peak intensity ratio, peak width, peak distance, and classical least squares can all be effective methods for the screening of excipients. Interesting excipient-dependent phase transformation behavior was also observed for probucol.
Collapse
Affiliation(s)
- Xiaoyun Chen
- Analytical Sciences, Core R&D, Dow Chemical Company, Midland, MI 48667 USA
| | | | | | | | | |
Collapse
|
28
|
Hoffmann M, Wray PS, Gamble JF, Tobyn M. Investigation into process-induced de-aggregation of cohesive micronised API particles. Int J Pharm 2015; 493:341-6. [DOI: 10.1016/j.ijpharm.2015.07.073] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 07/23/2015] [Accepted: 07/28/2015] [Indexed: 11/29/2022]
|
29
|
Paudel A, Raijada D, Rantanen J. Raman spectroscopy in pharmaceutical product design. Adv Drug Deliv Rev 2015; 89:3-20. [PMID: 25868453 DOI: 10.1016/j.addr.2015.04.003] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 03/15/2015] [Accepted: 04/01/2015] [Indexed: 12/20/2022]
Abstract
Almost 100 years after the discovery of the Raman scattering phenomenon, related analytical techniques have emerged as important tools in biomedical sciences. Raman spectroscopy and microscopy are frontier, non-invasive analytical techniques amenable for diverse biomedical areas, ranging from molecular-based drug discovery, design of innovative drug delivery systems and quality control of finished products. This review presents concise accounts of various conventional and emerging Raman instrumentations including associated hyphenated tools of pharmaceutical interest. Moreover, relevant application cases of Raman spectroscopy in early and late phase pharmaceutical development, process analysis and micro-structural analysis of drug delivery systems are introduced. Finally, potential areas of future advancement and application of Raman spectroscopic techniques are discussed.
Collapse
|
30
|
Potter C, Tian Y, Walker G, McCoy C, Hornsby P, Donnelly C, Jones DS, Andrews GP. Novel Supercritical Carbon Dioxide Impregnation Technique for the Production of Amorphous Solid Drug Dispersions: A Comparison to Hot Melt Extrusion. Mol Pharm 2015; 12:1377-90. [DOI: 10.1021/mp500644h] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | | | - Gavin Walker
- Department
of Chemical and Environmental Science, University of Limerick, Castletroy, Co. Limerick, Ireland
| | | | | | | | | | | |
Collapse
|
31
|
Ueda H, Wakabayashi S, Kikuchi J, Ida Y, Kadota K, Tozuka Y. Anomalous role change of tertiary amino and ester groups as hydrogen acceptors in eudragit E based solid dispersion depending on the concentration of naproxen. Mol Pharm 2015; 12:1050-61. [PMID: 25654583 DOI: 10.1021/mp5005417] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Eudragit E (EGE) is a basic polymer incorporating tertiary amino and ester groups. The role of the functional groups of EGE in the formation of solid dispersion (SD) with Naproxen (NAP) was investigated. The glass transition temperature (Tg) of EGE decreased with the plasticizing effect of NAP up to 20% weight ratio. Addition of NAP at over 30% induced a rise in Tg, with the maximum value being reached at 60% NAP. Further addition of NAP led to a rapid drop of the Tg. A dramatic difference of physical stability between the SDs including 60 and 70% NAP was confirmed. The SD including 70% NAP rapidly crystallized at 40 °C with 75% relative humidity, while the amorphous state could be maintained over 6 months in the SD with 60% NAP. The infrared and (13)C solid state-NMR spectra of the SDs suggested a formation of ionic interaction between the carboxylic acid of NAP and the amino group of EGE. The SD with 20% NAP raised the (13)C spin-lattice relaxation (T1) of the amino group, but it decreased with over 30% NAP. The change in the (13)C-T1 disappeared with 70% NAP. The (13)C-T1 of the ester group rose depending on the amount of NAP. From these findings, we concluded that the role as hydrogen acceptor shifted from the amine to the ester group with an increase in amount of NAP. Furthermore, the amino group of EGE did not contribute to the interaction at over 70% NAP. These phenomena could be strongly correlated with Tg and stability.
Collapse
Affiliation(s)
- Hiroshi Ueda
- ‡Laboratory of Formulation Design and Pharmaceutical Technology, Osaka University of Pharmaceutical Sciences, 4-20-1, Nasahara, Takatsuki-shi, Osaka 569-1094, Japan
| | | | | | | | - Kazunori Kadota
- ‡Laboratory of Formulation Design and Pharmaceutical Technology, Osaka University of Pharmaceutical Sciences, 4-20-1, Nasahara, Takatsuki-shi, Osaka 569-1094, Japan
| | - Yuichi Tozuka
- ‡Laboratory of Formulation Design and Pharmaceutical Technology, Osaka University of Pharmaceutical Sciences, 4-20-1, Nasahara, Takatsuki-shi, Osaka 569-1094, Japan
| |
Collapse
|
32
|
Gala U, Chauhan H. Principles and applications of Raman spectroscopy in pharmaceutical drug discovery and development. Expert Opin Drug Discov 2014; 10:187-206. [DOI: 10.1517/17460441.2015.981522] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Urvi Gala
- 1Creighton University, School of Pharmacy and Health Professions, 2500 California Plaza, Omaha, NE 68178, USA
| | - Harsh Chauhan
- 2Creighton University, School of Pharmacy and Health Professions, Department of Pharmacy Sciences, 2500 California Plaza, Omaha, NE 68178, USA ;
| |
Collapse
|
33
|
Rahman Z, Siddiqui A, Bykadi S, Khan MA. Determination of tacrolimus crystalline fraction in the commercial immediate release amorphous solid dispersion products by a standardized X-ray powder diffraction method with chemometrics. Int J Pharm 2014; 475:462-70. [DOI: 10.1016/j.ijpharm.2014.08.050] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 08/21/2014] [Accepted: 08/26/2014] [Indexed: 11/29/2022]
|
34
|
Onuki Y, Hasegawa N, Ikegami-Kawai M, Suematsu T, Sakurai S, Shirozu S, Tsubuki M, Obata Y, Takayama K. Contribution of glucose to crystallization of phenytoin in injectable dosage form by dilution with infusion fluids. Chem Pharm Bull (Tokyo) 2014; 62:989-93. [PMID: 25273057 DOI: 10.1248/cpb.c14-00353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The crystallization of phenytoin occurring after its dilution with infusion fluid is a major concern in the clinical use of injectable phenytoin. To gain further understanding of the crystallization, this study assessed details of the involvement of glucose in this action. For sample preparation, phenytoin crystals were created by diluting the injectable phenytoin with infusion fluids with different glucose concentrations at different temperature, and then the characteristics of the crystallization (e.g., crystal size in the long direction, accumulated amount over 24 h, and crystallization rate constant) were measured. Results of the analysis of variance indicated that the glucose concentration and temperature had significant impacts on the crystallization. The mode of action of the glucose concentration was suggested to be different from that of the incubation temperature. This study also examined the molecular mobility of components (i.e., glucose, propylene glycol, phenytoin) in the admixtures using diffusion NMR techniques. The findings will provide valuable information for the clinical use of injectable phenytoin.
Collapse
|
35
|
Scoutaris N, Vithani K, Slipper I, Chowdhry B, Douroumis D. SEM/EDX and confocal Raman microscopy as complementary tools for the characterization of pharmaceutical tablets. Int J Pharm 2014; 470:88-98. [DOI: 10.1016/j.ijpharm.2014.05.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/08/2014] [Accepted: 05/10/2014] [Indexed: 11/24/2022]
|
36
|
Brown C, DiNunzio J, Eglesia M, Forster S, Lamm M, Lowinger M, Marsac P, McKelvey C, Meyer R, Schenck L, Terife G, Troup G, Smith-Goettler B, Starbuck C. Hot-Melt Extrusion for Solid Dispersions: Composition and Design Considerations. ADVANCES IN DELIVERY SCIENCE AND TECHNOLOGY 2014. [DOI: 10.1007/978-1-4939-1598-9_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
37
|
Lu Y, Sturek M, Park K. Microparticles produced by the hydrogel template method for sustained drug delivery. Int J Pharm 2013; 461:258-69. [PMID: 24333903 DOI: 10.1016/j.ijpharm.2013.11.058] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 10/17/2013] [Accepted: 11/30/2013] [Indexed: 11/27/2022]
Abstract
Polymeric microparticles have been used widely for sustained drug delivery. Current methods of microparticle production can be improved by making homogeneous particles in size and shape, increasing the drug loading, and controlling the initial burst release. In the current study, the hydrogel template method was used to produce homogeneous poly(lactide-co-glycolide) (PLGA) microparticles and to examine formulation and process-related parameters. Poly(vinyl alcohol) (PVA) was used to make hydrogel templates. The parameters examined include PVA molecular weight, type of PLGA (as characterized by lactide content, inherent viscosity), polymer concentration, drug concentration and composition of solvent system. Three model compounds studied were risperidone, methylprednisolone acetate and paclitaxel. The ability of the hydrogel template method to produce microparticles with good conformity to template was dependent on molecular weight of PVA and viscosity of the PLGA solution. Drug loading and encapsulation efficiency were found to be influenced by PLGA lactide content, polymer concentration and composition of the solvent system. The drug loading and encapsulation efficiency were 28.7% and 82% for risperidone, 31.5% and 90% for methylprednisolone acetate, and 32.2% and 92% for paclitaxel, respectively. For all three drugs, release was sustained for weeks, and the in vitro release profile of risperidone was comparable to that of microparticles prepared using the conventional emulsion method. The hydrogel template method provides a new approach of manipulating microparticles.
Collapse
Affiliation(s)
- Ying Lu
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN 47906, USA
| | - Michael Sturek
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kinam Park
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN 47906, USA; Department of Biomedical Engineering, Purdue University, West Lafayette, IN 47906, USA.
| |
Collapse
|
38
|
Vogt FG, Strohmeier M. Confocal UV and Resonance Raman Microscopic Imaging of Pharmaceutical Products. Mol Pharm 2013; 10:4216-28. [DOI: 10.1021/mp400314s] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Frederick G. Vogt
- Product
Development, GlaxoSmithKline plc., 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| | - Mark Strohmeier
- Product
Development, GlaxoSmithKline plc., 709 Swedeland Road, King of Prussia, Pennsylvania 19406, United States
| |
Collapse
|
39
|
Guo Y, Shalaev E, Smith S. Physical stability of pharmaceutical formulations: solid-state characterization of amorphous dispersions. Trends Analyt Chem 2013. [DOI: 10.1016/j.trac.2013.06.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
40
|
Investigation of polymeric excipients for dutasteride solid dispersion and its physicochemical characterization. Arch Pharm Res 2013; 37:214-24. [DOI: 10.1007/s12272-013-0180-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 06/04/2013] [Indexed: 11/25/2022]
|
41
|
Paudel A, Loyson Y, Van den Mooter G. An Investigation into the Effect of Spray Drying Temperature and Atomizing Conditions on Miscibility, Physical Stability, and Performance of Naproxen–PVP K 25 Solid Dispersions. J Pharm Sci 2013; 102:1249-67. [DOI: 10.1002/jps.23459] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 12/17/2012] [Accepted: 01/09/2013] [Indexed: 11/11/2022]
|
42
|
Impact of Excipient Interactions on Solid Dosage Form Stability. Pharm Res 2012; 29:2660-83. [DOI: 10.1007/s11095-012-0782-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 05/14/2012] [Indexed: 10/28/2022]
|