1
|
Sacher S, Fink E, Alva C, Alberto Afonso Urich J, Doğan A, Herndler V, Koutsamanis I, Kushwah V, Peter A, Salar-Behzadi S, Wilfling K, Stranzinger S, Zettl M, Feng X, Korang-Yeboah M, Wu H, Khinast JG. Real-time prediction of dissolution profiles of coated oral dosage forms. Int J Pharm 2024; 666:124841. [PMID: 39414187 DOI: 10.1016/j.ijpharm.2024.124841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/03/2024] [Accepted: 10/14/2024] [Indexed: 10/18/2024]
Abstract
Optical coherence tomography (OCT) has emerged as an in-line monitoring technique for pharmaceutical coating processes based on a representative number of samples. In this study, an approach was developed to correlate the coating thickness measured in-line via OCT with the resultant tablet dissolution profile. This strategy enables prediction of the dissolution profile of coated oral dosage forms for each individual state of the coating process in real-time. Correlation models were developed for a tablet pan coating process and for a pellet fluid-bed coating process. The feasibility of the correlation models was tested using different process parameters and types of coating formulations. This work demonstrated that using the OCT data to predict dissolution could possibly form a unique way of assuring drug product quality and establishing a control strategy within the real-time release testing (RTRT) concept.
Collapse
Affiliation(s)
- Stephan Sacher
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010 Graz, Austria.
| | - Elisabeth Fink
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010 Graz, Austria
| | - Carolina Alva
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010 Graz, Austria
| | | | - Aygün Doğan
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010 Graz, Austria
| | - Vanessa Herndler
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010 Graz, Austria
| | - Ioannis Koutsamanis
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010 Graz, Austria
| | - Varun Kushwah
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010 Graz, Austria
| | - Anna Peter
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010 Graz, Austria
| | | | - Katrina Wilfling
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010 Graz, Austria
| | - Sandra Stranzinger
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010 Graz, Austria
| | - Manuel Zettl
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010 Graz, Austria
| | - Xin Feng
- United States Food and Drug Administration, Silver Spring, MD 20993, United States
| | | | - Huiquan Wu
- United States Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Johannes G Khinast
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010 Graz, Austria; Institute for Process and Particle Engineering, Graz University of Technology, Inffeldgasse 13/3, 8010 Graz, Austria
| |
Collapse
|
2
|
Dias RC, Korhonen O, Ketolainen J, Ervasti T, Lopes JA. The effect of process parameters on a continuous blending process monitored in-line by near-infrared spectroscopy. Eur J Pharm Sci 2024; 202:106890. [PMID: 39214317 DOI: 10.1016/j.ejps.2024.106890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/23/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
The continuous feeding-mixing system ensures the composition uniformity down to the tableting continuous manufacturing line so that a quality end-product is consistently delivered. Near-infrared spectroscopy (NIRS) enables in-line assessment of the blend's critical quality attributes in real-time. In this study, the effect of total feed rate and impeller speed on the continuous blending process monitored in-line by NIRS was examined by principal component analysis (PCA), ANOVA simultaneous component analysis (ASCA) and partial least squares (PLS) regression. Process data were generated by a factorial experimental design with process parameters and a constant formulation comprised of: 30 % (wt/wt) ibuprofen, 67.5 % (wt/wt) microcrystalline cellulose, 2 % (wt/wt) of sodium starch glycolate and 0.5 % (wt/wt) of magnesium stearate. The PCA hinted at the prevalence of impeller speed effect on ibuprofen concentration due to path length variation of the NIR light caused by the fluidized behaviour in the powder blend as a result of high speed ranges (>300 rpm). The ASCA model indicated that while both impeller speed and total feed rate effects were statistically significant (p-value=0.004), the impeller speed was the factor that contributed the most to the spectral variance (55.5 %). The PLS regression model for the ibuprofen content resulted in a RMSECV of 1.3 % (wt/wt) and showed that impeller speed was yet again the factor that exerted the major influence on spectral variance, owing to its wavelength-dependent effect that prevents common pre-processing techniques from eliminating it across the entire NIR range. The best sample presentation to the NIR probe was achieved at low impeller speed ranges (<600 rpm) and low total feed rates (<15 kg/h), such that it enhanced the PLS model ability to predict the ibuprofen concentration in the blend.
Collapse
Affiliation(s)
- Rute C Dias
- PromisLab, School of Pharmacy, University of Eastern Finland, 70211 Kuopio, Finland; iMed.ULisboa, Faculty of Pharmacy, University of Lisbon, 1649-003 Lisbon, Portugal.
| | - Ossi Korhonen
- PromisLab, School of Pharmacy, University of Eastern Finland, 70211 Kuopio, Finland
| | - Jarkko Ketolainen
- PromisLab, School of Pharmacy, University of Eastern Finland, 70211 Kuopio, Finland
| | - Tuomas Ervasti
- PromisLab, School of Pharmacy, University of Eastern Finland, 70211 Kuopio, Finland
| | - João A Lopes
- iMed.ULisboa, Faculty of Pharmacy, University of Lisbon, 1649-003 Lisbon, Portugal
| |
Collapse
|
3
|
Rangel-Gil RS, Nasrala-Álvarez JM, Romañach RJ, Méndez R. Blend uniformity monitoring in a continuous manufacturing mixing process for a low-dosage formulation using a stream sampler and near infrared spectroscopy. Int J Pharm 2024; 661:124478. [PMID: 39019300 DOI: 10.1016/j.ijpharm.2024.124478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/08/2024] [Accepted: 07/13/2024] [Indexed: 07/19/2024]
Abstract
Continuous manufacturing has the potential to offer several benefits for the production of oral solid dosage forms, including reduced costs, low-scale equipment, and the application of process analytical technology (PAT) for real-time process control. This study focuses on the implementation of a stream sampler to develop a near infrared (NIR) calibration model for blend uniformity monitoring in a continuous manufacturing mixing process. Feeding and mixing characterizations were performed for three loss-in-weight feeders and a commercial continuous mixer to prepare powder blends of 2.5-7.5 % w/w ibuprofen DC 85 W with a total throughput of 33 kg/h. The NIR spectral acquisition was performed after the mixing stage using a stream sampler for flowing powders. A continuous mixer shaft speed of 250 RPM was selected to operate the mixing process based on a variability analysis developed with in-line spectral data acquired using the stream sampler at 6 RPM. A partial least squares regression (PLS-R) model was performed and evaluated, yielding a root-mean-square error of prediction (RMSEP) of 0.39 % w/w and a bias of 0.05 % w/w. An independent experimental run conducted two days later revealed that the continuous mixing process and the NIR calibration model presented low day-to-day variation. The minimum practical error (MPE) and sill values through variographic analysis showed low variance associated with the sampling process using the stream sampler. Results demonstrated the promising capacity of the stream sampler coupled to an NIR probe to be implemented within continuous manufacturing processes for the real-time determination of API concentration.
Collapse
Affiliation(s)
- Raúl S Rangel-Gil
- Department of Chemical Engineering, University of Puerto Rico at Mayaguez, Puerto Rico 00681, United States
| | - Juan M Nasrala-Álvarez
- Department of Chemical Engineering, University of Puerto Rico at Mayaguez, Puerto Rico 00681, United States
| | - Rodolfo J Romañach
- Department of Chemistry, University of Puerto Rico at Mayaguez, Puerto Rico 00681, United States
| | - Rafael Méndez
- Department of Chemical Engineering, University of Puerto Rico at Mayaguez, Puerto Rico 00681, United States.
| |
Collapse
|
4
|
Sacher S, Kottlan A, Diop JB, Heimsten R. Prediction of in-vitro dissolution and tablet hardness from optical porosity measurements. Int J Pharm 2024; 660:124336. [PMID: 38871136 DOI: 10.1016/j.ijpharm.2024.124336] [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: 05/08/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/15/2024]
Abstract
Advanced manufacturing technologies such as continuous processing require fast information on the quality of intermediates and products. Process analytical technologies (PAT) to monitor many critical quality attributes (CQAs) have been developed and successfully implemented in pharmaceutical industry. However, there are some CQAs, which still have to be measured off-line with significant effort due to the lack of suitable PAT sensors. Two prominent examples are the in-vitro dissolution and the tablet hardness. Both are obtained via destructive measurement, and the dissolution is tedious and time-consuming to determine. In this study, these two CQAs were predicted via correlation with the optical porosity of tablets. The optical porosity was measured via a novel combination of gas in scattering media absorption spectroscopy (GASMAS) and photon time of flight spectroscopy (pTOFS) with a SpectraPore instrument. The approach was tested in a continuous tableting line and showed promising results in predicting the amount of drug released after specific dissolution times as well as the tablet hardness. This indicates that the measurement of optical porosity can support control strategies within the real-time release testing (RTRT) concept.
Collapse
Affiliation(s)
- Stephan Sacher
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2 8010, Graz, Austria.
| | - Andreas Kottlan
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2 8010, Graz, Austria
| | | | | |
Collapse
|
5
|
Janssen PHM, Fathollahi S, Dickhoff BHJ, Frijlink HW. Critical review on the role of excipient properties in pharmaceutical powder-to-tablet continuous manufacturing. Expert Opin Drug Deliv 2024; 21:1069-1079. [PMID: 39129595 DOI: 10.1080/17425247.2024.2384698] [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: 05/15/2024] [Accepted: 07/22/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION The pharmaceutical industry is gradually changing batch-wise manufacturing processes to continuous manufacturing processes, due to the advantages it has to offer. The final product quality and process efficiency of continuous manufacturing processes is among others impacted by the properties of the raw materials. Existing knowledge on the role of raw material properties in batch processing is however not directly transferable to continuous processes, due to the inherent differences between batch and continuous processes. AREAS COVERED A review is performed to evaluate the role of excipient properties for different unit operations used in continuous manufacturing processes. Unit operations that will be discussed include feeding, blending, granulation, final blending, and compression. EXPERT OPINION Although the potency of continuous manufacturing is widely recognized, full utilization still requires a number of challenges to be addressed effectively. An expert opinion will be provided that discusses those challenges and potential solutions to overcome those challenges. The provided overview can serve as a framework for the pharmaceutical industry to push ahead process optimization and formulation development for continuous manufacturing processes.
Collapse
Affiliation(s)
- Pauline H M Janssen
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, The Netherlands
- Innovation & Technical Solutions, DFE Pharma, Goch, Germany
| | | | | | - Henderik W Frijlink
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
6
|
Denduyver P, Birk G, Ambruosi A, Vervaet C, Vanhoorne V. Evaluation of Polyvinyl Alcohol as Binder during Continuous Twin Screw Wet Granulation. Pharmaceutics 2024; 16:854. [PMID: 39065551 PMCID: PMC11280237 DOI: 10.3390/pharmaceutics16070854] [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: 05/23/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024] Open
Abstract
Binder selection is a crucial step in continuous twin-screw wet granulation (TSWG), as the material experiences a much shorter residence time (2-40 s) in the granulator barrel compared to batch-wise granulation processes. Polyvinyl alcohol (PVA) 4-88 was identified as an effective binder during TSWG, but the potential of other PVA grades-differing in polymerization and hydrolysis degree-has not yet been studied. Therefore, the aim of the current study was to evaluate the potential of different PVA grades as a binder during TSWG. The breakage and drying behavior during the fluidized bed drying of drug-loaded granules containing the PVA grades was also studied. Three PVA grades (4-88, 18-88, and 40-88) were characterized and their attributes were compared to previously investigated binders by Vandevivere et al. through principal component analysis. Three binder clusters could be distinguished according to their attributes, whereby each cluster contained a PVA grade and a previously investigated binder. PVA 4-88 was the most effective binder of the PVA grades for both a good water-soluble and water-insoluble formulation. This could be attributed to its high total surface energy, low viscosity, good wettability of hydrophilic and hydrophobic surfaces, and good wettability by water of the binder. Compared to the previously investigated binders, all PVA grades were more effective in the water-insoluble formulation, as they yielded strong granules (friability below 30%) at lower L/S-ratios. This was linked to the high dispersive surface energy of the high-energy sites on the surface of PVA grades and their low surface tension. During fluidized bed drying, PVA grades proved suitable binders, as the acetaminophen (APAP) granules were dried within a short time due to the low L/S-ratio, at which high-quality granules could be produced. In addition, no attrition occurred, and strong tablets were obtained. Based on this study, PVA could be the preferred binder during twin screw granulation due to its high binder effectiveness at a low L/S-ratio, allowing efficient downstream processing. However, process robustness must be controlled by the included excipients, as PVA grades are operating in a narrow L/S-ratio range.
Collapse
Affiliation(s)
- Phaedra Denduyver
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; (P.D.); (C.V.)
| | - Gudrun Birk
- Merck KGaA, Frankfuter Str. 250, 64293 Darmstadt, Germany; (G.B.); (A.A.)
| | | | - Chris Vervaet
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; (P.D.); (C.V.)
| | - Valérie Vanhoorne
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; (P.D.); (C.V.)
| |
Collapse
|
7
|
Lyytikäinen J, Stasiak P, Kubelka T, Bogaerts I, Wanek A, Stynen B, Holman J, Ketolainen J, Ervasti T, Korhonen O. Continuous direct compression of a commercially batch-manufactured tablet formulation with two different processing lines. Eur J Pharm Biopharm 2024; 199:114278. [PMID: 38583787 DOI: 10.1016/j.ejpb.2024.114278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/05/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
The transfer from batch-based to continuous tablet manufacturing increases the quality and efficiency of processes. Nonetheless, as in the development of a batch process, the continuous process design requires optimization studies to ensure a robust process. In this study, processing of a commercially batch-manufactured tablet product was tested with two continuous direct compression lines while keeping the original formulation composition and tablet quality requirements. Tableting runs were conducted with different values of process parameters. Changes in parameter settings were found to cause differences in tablet properties. Most of these quality properties could be controlled and maintained within the set limits effortlessly already at this stage of studies. However, the API content and content uniformity seemed to require more investigation. The observed content uniformity challenges were traced to individual tablets with a high amount of API. This was suspected to be caused by API micro-agglomerates since tablet weight variability did not explain the issue. This could be solved by adding a mill between two blenders in the process line. Overall, this case study produced promising results with both tested manufacturing lines since many tablet properties complied with the test result limits without optimization of process parameter settings.
Collapse
Affiliation(s)
- Jenna Lyytikäinen
- School of Pharmacy, PromisLab, University of Eastern Finland, Kuopio, Finland.
| | | | | | | | - Adam Wanek
- Zentiva, Prague, Czech Republic; UCT Prague, Prague, Czech Republic.
| | - Bart Stynen
- GEA Process Engineering, Wommelgem, Belgium.
| | | | - Jarkko Ketolainen
- School of Pharmacy, PromisLab, University of Eastern Finland, Kuopio, Finland.
| | - Tuomas Ervasti
- School of Pharmacy, PromisLab, University of Eastern Finland, Kuopio, Finland.
| | - Ossi Korhonen
- School of Pharmacy, PromisLab, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
8
|
Macchietti L, Melucci D, Menarini L, Consoli F, Zappi A. Analytical comparison between batch and continuous direct compression processes for pharmaceutical manufacturing using an innovative UV-Vis reflectance method and chemometrics. Int J Pharm 2024; 656:124090. [PMID: 38582101 DOI: 10.1016/j.ijpharm.2024.124090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
Advancements in industrial technologies and the application of quality by design (QbD) guidelines are shifting the attention of manufacturers towards innovative production techniques. In the pharmaceutical field, there is a significant focus on the implementation of continuous processes, in which the production stages are carried out continuously, without the need to interrupt the process and store the production intermediates, as in traditional batch production. Such innovative production techniques also require the development of proper analytical methods able to analyze the products in-line, while still being processed. The present study aims to compare a traditional batch manufacturing process with an alternative continuous one. To this end, a real pharmaceutical formulation was used, substituting the active pharmaceutical ingredient (API) with riboflavin, at the concentration of 2 %w/w. Moreover, a direct and non-destructive analytical method based on UV-Vis reflectance spectroscopy was applied for the quantification of riboflavin in the final tablets, and compared with a traditional absorbance analysis. Good results were obtained in the comparison of both the two manufacturing processes and the two analytical methods, with R2 higher than 0.9 for all the calculated calibration models and predicted riboflavin concentrations that never significantly overcame the 15 % limits recommended by the pharmacopeia. The continuous production method demonstrated to be as reliable as the batch one, allowing to save time and money in the production step. Moreover, UV-Vis reflectance was proved to be an interesting alternative to absorption spectroscopy, which, with the proper technology, could be implemented for in-line process control.
Collapse
Affiliation(s)
- Laura Macchietti
- Department of Chemistry "G. Ciamician", University of Bologna, 40126 Bologna, Italy.
| | - Dora Melucci
- Department of Chemistry "G. Ciamician", University of Bologna, 40126 Bologna, Italy.
| | | | | | - Alessandro Zappi
- Department of Chemistry "G. Ciamician", University of Bologna, 40126 Bologna, Italy.
| |
Collapse
|
9
|
Peeters M, Barrera Jiménez AA, Matsunami K, Ghijs M, Dos Santos Schultz E, Roudgar M, Vigh T, Stauffer F, Nopens I, De Beer T. Analysis of the effect of formulation properties and process parameters on granule formation in twin-screw wet granulation. Int J Pharm 2024; 650:123671. [PMID: 38065345 DOI: 10.1016/j.ijpharm.2023.123671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 12/01/2023] [Accepted: 12/02/2023] [Indexed: 01/08/2024]
Abstract
In the last few years, twin-screw wet granulation (TSWG) has become one of the key continuous pharmaceutical unit operations. Despite the many studies that have been performed, only little is known about the effect of the starting material properties on the stepwise granule formation along the length of the twin-screw granulator (TSG) barrel. Hence, this study obtained a detailed understanding of the effect of formulation properties (i.e., Active Pharmaceutical Ingredient (API) properties, formulation blend particle size distribution and formulation drug load) and process settings on granule formation in TSWG. An experimental set-up was used allowing the collection of granules at the different TSG compartments. Granules were characterized in terms of granule size, shape, binder liquid and API distributions. Liquid-to-solid (L/S) ratio was the only TSG process parameter impacting the granule size and shape evolution. Particle size and flow properties (e.g., flow rate index) had an important effect on the granule size and shape changes whereas water-related properties (e.g., water binding capacity and solubility) became influential at the last TSG compartments. The API solubility and L/S ratio were found to have a major impact on the distribution of binder liquid over the different granule size fractions. In the first TSG compartment (i.e., wetting compartment), the distribution of the API in the granules was influenced by its solubility in the granulation liquid.
Collapse
Affiliation(s)
- Michiel Peeters
- Laboratory of Pharmaceutical Process Analytical Technology, Department of Pharmaceutical Analysis, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Oost-Vlaanderen, Belgium
| | - Ana Alejandra Barrera Jiménez
- Laboratory of Pharmaceutical Process Analytical Technology, Department of Pharmaceutical Analysis, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Oost-Vlaanderen, Belgium; BIOMATH, Department of Data Analysis and Mathematical Modelling, Ghent University, Coupure links 653, Ghent 9000, Oost-Vlaanderen, Belgium
| | - Kensaku Matsunami
- Laboratory of Pharmaceutical Process Analytical Technology, Department of Pharmaceutical Analysis, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Oost-Vlaanderen, Belgium; BIOMATH, Department of Data Analysis and Mathematical Modelling, Ghent University, Coupure links 653, Ghent 9000, Oost-Vlaanderen, Belgium.
| | - Michael Ghijs
- Laboratory of Pharmaceutical Process Analytical Technology, Department of Pharmaceutical Analysis, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Oost-Vlaanderen, Belgium; BIOMATH, Department of Data Analysis and Mathematical Modelling, Ghent University, Coupure links 653, Ghent 9000, Oost-Vlaanderen, Belgium
| | | | - Mina Roudgar
- Discovery, Product Development & Supply, Janssen R&D, Beerse B-2340, Belgium
| | - Tamas Vigh
- Discovery, Product Development & Supply, Janssen R&D, Beerse B-2340, Belgium
| | - Fanny Stauffer
- Product Design & Performance, UCB, Braine l'Alleud 1420, Belgium
| | - Ingmar Nopens
- BIOMATH, Department of Data Analysis and Mathematical Modelling, Ghent University, Coupure links 653, Ghent 9000, Oost-Vlaanderen, Belgium
| | - Thomas De Beer
- Laboratory of Pharmaceutical Process Analytical Technology, Department of Pharmaceutical Analysis, Ghent University, Ottergemsesteenweg 460, Ghent 9000, Oost-Vlaanderen, Belgium
| |
Collapse
|
10
|
Kuchler L, Spoerk M, Eder S, Doğan A, Khinast J, Sacher S. Liquid API feeding in pharmaceutical HME: Novel options in solid dosage manufacturing. Int J Pharm 2024; 650:123690. [PMID: 38081563 DOI: 10.1016/j.ijpharm.2023.123690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023]
Abstract
Hot melt extrusion (HME) is a common unit operation. It is broadly applicable in the pharmaceutical industry and can be implemented in a continuous manufacturing line. However, the conventional way of active pharmaceutical ingredient (API) feeding with a pre-blend consisting of a powdered API and a polymer does not allow the flexibility and agility to adjust the process parameters, which is generally an essential part of continuous manufacturing. In addition, this method of API feeding may result in the segregation of the individual powder components or agglomeration of highly cohesive materials, leading to an inhomogeneous API content in the extrudates, especially at low doses. In this study, the universal applicability of liquid side feeding in pharmaceutical HME was demonstrated using various APIs suspended or dissolved in water and fed as suspension or undersaturated, supersaturated, and highly concentrated solutions into anterior parts of the extruder. The extrudates were characterized in terms of their API content, residual moisture content, and solid-state of the API embedded in the polymer. The results show that a uniform API content without major deviations can be obtained via this method. Furthermore, the residual moisture content of the extrudates was low enough to have no significant influence on further processing of the final dosage form. In summary, this advanced way of feeding allows an accurate, flexible, and agile feeding of APIs, facilitating the production of personalized final dosage forms and a novel option to link the manufacturing of the drug substance and the drug product.
Collapse
Affiliation(s)
- Lisa Kuchler
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria
| | - Martin Spoerk
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria; Institute for Process and Particle Engineering, Graz University of Technology, Inffeldgasse 13/3, 8010 Graz, Austria
| | - Simone Eder
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria
| | - Aygün Doğan
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria
| | - Johannes Khinast
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria; Institute for Process and Particle Engineering, Graz University of Technology, Inffeldgasse 13/3, 8010 Graz, Austria
| | - Stephan Sacher
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13, 8010 Graz, Austria.
| |
Collapse
|
11
|
Velez-Silva NL, Drennen JK, Anderson CA. Continuous manufacturing of pharmaceutical products: A density-insensitive near infrared method for the in-line monitoring of continuous powder streams. Int J Pharm 2024; 650:123699. [PMID: 38081558 DOI: 10.1016/j.ijpharm.2023.123699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
Near infrared (NIR) spectroscopy is a valuable analytical technique for monitoring chemical composition of powder blends in continuous pharmaceutical processes. However, the variation in density captured by NIR during spectral collection of dynamic powder streams at different flow rates often reduces the performance and robustness of NIR models. To overcome this challenge, quantitative NIR measurements are commonly collected across all potential manufacturing conditions, including multiple flow rates to account for the physical variations. The utility of this approach is limited by the considerable quantity of resources required to run and analyze an extensive calibration design at variable flow rates in a continuous manufacturing (CM) process. It is hypothesized that the primary variation introduced to NIR spectra from changing flow rates is a change in the density of the powder from which NIR spectra are collected. In this work, powder stream density was used as an efficient surrogate for flow rate in developing a quantitative NIR method with enhanced robustness against process rate variation. A density design space of two process parameters was generated to determine the conditions required to encompass the apparent density and spectral variance from increases in process rate. This apparent density variance was included in calibration at a constant low flow rate to enable the development of a density-insensitive NIR quantitative model with limited consumption of materials. The density-insensitive NIR model demonstrated comparable prediction performance and flow rate robustness to a traditional NIR model including flow rate variation ("gold standard" model) when applied to monitoring drug content in continuous runs at varying flow rates. The proposed platform for the development of in-line density-insensitive NIR methods is expected to facilitate robust analytical model performance across variable continuous manufacturing production scales while improving the material efficiency over traditional robust modeling approaches for calibration development.
Collapse
Affiliation(s)
- Natasha L Velez-Silva
- Duquesne University Graduate School for Pharmaceutical Sciences, Pittsburgh, PA 15282, United States; Duquesne Center for Pharmaceutical Technology, Duquesne University, Pittsburgh, PA 15282, United States
| | - James K Drennen
- Duquesne University Graduate School for Pharmaceutical Sciences, Pittsburgh, PA 15282, United States; Duquesne Center for Pharmaceutical Technology, Duquesne University, Pittsburgh, PA 15282, United States
| | - Carl A Anderson
- Duquesne University Graduate School for Pharmaceutical Sciences, Pittsburgh, PA 15282, United States; Duquesne Center for Pharmaceutical Technology, Duquesne University, Pittsburgh, PA 15282, United States.
| |
Collapse
|
12
|
Sun S, Alkahtani ME, Gaisford S, Basit AW, Elbadawi M, Orlu M. Virtually Possible: Enhancing Quality Control of 3D-Printed Medicines with Machine Vision Trained on Photorealistic Images. Pharmaceutics 2023; 15:2630. [PMID: 38004607 PMCID: PMC10674815 DOI: 10.3390/pharmaceutics15112630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Three-dimensional (3D) printing is an advanced pharmaceutical manufacturing technology, and concerted efforts are underway to establish its applicability to various industries. However, for any technology to achieve widespread adoption, robustness and reliability are critical factors. Machine vision (MV), a subset of artificial intelligence (AI), has emerged as a powerful tool to replace human inspection with unprecedented speed and accuracy. Previous studies have demonstrated the potential of MV in pharmaceutical processes. However, training models using real images proves to be both costly and time consuming. In this study, we present an alternative approach, where synthetic images were used to train models to classify the quality of dosage forms. We generated 200 photorealistic virtual images that replicated 3D-printed dosage forms, where seven machine learning techniques (MLTs) were used to perform image classification. By exploring various MV pipelines, including image resizing and transformation, we achieved remarkable classification accuracies of 80.8%, 74.3%, and 75.5% for capsules, tablets, and films, respectively, for classifying stereolithography (SLA)-printed dosage forms. Additionally, we subjected the MLTs to rigorous stress tests, evaluating their scalability to classify over 3000 images and their ability to handle irrelevant images, where accuracies of 66.5% (capsules), 72.0% (tablets), and 70.9% (films) were obtained. Moreover, model confidence was also measured, and Brier scores ranged from 0.20 to 0.40. Our results demonstrate promising proof of concept that virtual images exhibit great potential for image classification of SLA-printed dosage forms. By using photorealistic virtual images, which are faster and cheaper to generate, we pave the way for accelerated, reliable, and sustainable AI model development to enhance the quality control of 3D-printed medicines.
Collapse
Affiliation(s)
- Siyuan Sun
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (S.S.); (M.E.A.); (S.G.)
| | - Manal E. Alkahtani
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (S.S.); (M.E.A.); (S.G.)
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Simon Gaisford
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (S.S.); (M.E.A.); (S.G.)
| | - Abdul W. Basit
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (S.S.); (M.E.A.); (S.G.)
| | - Moe Elbadawi
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (S.S.); (M.E.A.); (S.G.)
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4DQ, UK
| | - Mine Orlu
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (S.S.); (M.E.A.); (S.G.)
| |
Collapse
|
13
|
Velez-Silva NL, Drennen JK, Anderson CA. Influence of powder stream density on near infrared measurements upon scale-up of a simulated continuous process. Int J Pharm 2023; 645:123354. [PMID: 37647977 DOI: 10.1016/j.ijpharm.2023.123354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/17/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023]
Abstract
Near-infrared (NIR) spectroscopy is a powerful process analytical tool for monitoring chemical constituents in continuous pharmaceutical processes. However, the density variation introduced when quantitative NIR measurements are performed on powder streams at different flow rates is a potential source of a lack of model robustness. Since different flow rates are often required to meet the production requirements (e.g., during scale-up) of a continuous process, the development of efficient strategies to characterize, understand, and mitigate the impact of powder density on NIR measurements is highly desirable. This study focused on assessing the effect of powder physical variation on NIR by enabling the in-line characterization of powder stream density in a simulated continuous system. The in-line measurements of powder stream density were facilitated through a unique analytical interface to a flowing process. Powder streams delivered at various design levels of flow rate and tube angle were monitored simultaneously by NIR diffuse reflectance spectroscopy, live imaging, and dynamic mass characterization. Statistical analysis and multivariate modeling confirmed powder density as a significant source of spectral variability due to flow rate. Besides providing broader process understanding, results elucidated potential mitigation strategies to facilitate effective continuous process scale-up while ensuring NIR model robustness against density.
Collapse
Affiliation(s)
- Natasha L Velez-Silva
- Duquesne University Graduate School for Pharmaceutical Sciences, Pittsburgh, PA 15282, United States; Duquesne Center for Pharmaceutical Technology, Duquesne University, Pittsburgh, PA 15282, United States
| | - James K Drennen
- Duquesne University Graduate School for Pharmaceutical Sciences, Pittsburgh, PA 15282, United States; Duquesne Center for Pharmaceutical Technology, Duquesne University, Pittsburgh, PA 15282, United States
| | - Carl A Anderson
- Duquesne University Graduate School for Pharmaceutical Sciences, Pittsburgh, PA 15282, United States; Duquesne Center for Pharmaceutical Technology, Duquesne University, Pittsburgh, PA 15282, United States.
| |
Collapse
|
14
|
Vasoya JM, Lee HL, Lee T, Serajuddin ATM. Continuous Synthesis of Cinnarizine Salt with Malic Acid by Applying Green Chemistry Using Water-Assisted Twin Screw Extrusion. Mol Pharm 2023; 20:5160-5172. [PMID: 37646101 DOI: 10.1021/acs.molpharmaceut.3c00511] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Organic solvent-free process or green chemistry is needed for manufacturing pharmaceutical salts to avoid various environmental, safety, and manufacturing cost issues involved. In this study, a cinnarizine (CNZ) salt with malic acid at a 1:1 molar ratio was successfully prepared by twin screw extrusion (TSE) with water assistance. The feasibility of salt formation was first evaluated by screening several carboxylic acids by neat grinding (NG) and liquid-assisted grinding (LAG) using a mortar and pestle, which indicated that malic acid and succinic acid could form salts with CNZ. Further studies on salt formation were conducted using malic acid. The examination by hot-stage microscopy revealed that the addition of water could facilitate the formation and crystallization of CNZ-malic acid salt even though CNZ is poorly water-soluble. The feasibility of salt formation was confirmed by determining the pH-solubility relationship between CNZ and malic acid, where a pHmax of 2.7 and a salt solubility of 2.47 mg/mL were observed. Authentic salt crystals were prepared by solution crystallization from organic solvents for examining crystal properties and structure by differential scanning calorimetry (DSC), powder X-ray diffraction (PXRD), Fourier transform infrared (FTIR) spectroscopy, solid-state 13C and 15N nuclear magnetic resonance (NMR), and single-crystal X-ray diffraction (SXD). These techniques also established that a salt, and not a cocrystal, was indeed formed. The CNZ salt crystals were then prepared by TSE of a 1:1 CNZ-malic acid mixture, where the addition of small amounts of water resulted in a complete conversion of the mixture into the salt form. The salts prepared by solvent crystallization and water-assisted TSE had identical properties, and their moisture sorption profiles were also similar, indicating that TSE is a viable method for salt preparation by green chemistry. Since TSE can be conducted in a continuous manner, the results of the present investigation, if combined with other continuous processes, suggest the possibility of continuous manufacturing of drug products from the synthesis of active pharmaceutical ingredients (APIs) to the production of final dosage forms.
Collapse
Affiliation(s)
- Jaydip M Vasoya
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York 11439, United States
| | - Hung Lin Lee
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York 11439, United States
- Department of Chemical and Materials Engineering, National Central University, 300 Zhongda Road, Zhongli District, Taoyuan City 320317, Taiwan, ROC
| | - Tu Lee
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York 11439, United States
- Department of Chemical and Materials Engineering, National Central University, 300 Zhongda Road, Zhongli District, Taoyuan City 320317, Taiwan, ROC
| | - Abu T M Serajuddin
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York 11439, United States
| |
Collapse
|
15
|
Péterfi O, Madarász L, Ficzere M, Lestyán-Goda K, Záhonyi P, Erdei G, Sipos E, Nagy ZK, Galata DL. In-line particle size measurement during granule fluidization using convolutional neural network-aided process imaging. Eur J Pharm Sci 2023; 189:106563. [PMID: 37582409 DOI: 10.1016/j.ejps.2023.106563] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/24/2023] [Accepted: 08/12/2023] [Indexed: 08/17/2023]
Abstract
This paper presents a machine learning-based image analysis method to monitor the particle size distribution of fluidized granules. The key components of the direct imaging system are a rigid fiber-optic endoscope, a light source and a high-speed camera, which allow for real-time monitoring of the granules. The system was implemented into a custom-made 3D-printed device that could reproduce the particle movement characteristic in a fluidized-bed granulator. The suitability of the method was evaluated by determining the particle size distribution (PSD) of various granule mixtures within the 100-2000 μm size range. The convolutional neural network-based software was able to successfully detect the granules that were in focus despite the dense flow of the particles. The volumetric PSDs were compared with off-line reference measurements obtained by dynamic image analysis and laser diffraction. Similar trends were observed across the PSDs acquired with all three methods. The results of this study demonstrate the feasibility of performing real-time particle size analysis using machine vision as an in-line process analytical technology (PAT) tool.
Collapse
Affiliation(s)
- Orsolya Péterfi
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| | - Lajos Madarász
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| | - Máté Ficzere
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| | - Katalin Lestyán-Goda
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| | - Petra Záhonyi
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| | - Gábor Erdei
- Department of Atomic Physics, Faculty of Natural Sciences, Budapest University of Technology and Economics, H-1111, Budapest, Budafoki 8, Hungary
| | - Emese Sipos
- Department of Pharmaceutical Industry and Management, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, Gheorghe Marinescu street 38, 540142 Targu Mures, Romania
| | - Zsombor Kristóf Nagy
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary.
| | - Dorián László Galata
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., H-1111 Budapest, Hungary
| |
Collapse
|
16
|
Galvan D, de Aguiar LM, Bona E, Marini F, Killner MHM. Successful combination of benchtop nuclear magnetic resonance spectroscopy and chemometric tools: A review. Anal Chim Acta 2023; 1273:341495. [PMID: 37423658 DOI: 10.1016/j.aca.2023.341495] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/20/2023] [Accepted: 06/05/2023] [Indexed: 07/11/2023]
Abstract
Low-field nuclear magnetic resonance (NMR) has three general modalities: spectroscopy, imaging, and relaxometry. In the last twelve years, the modality of spectroscopy, also known as benchtop NMR, compact NMR, or just low-field NMR, has undergone instrumental development due to new permanent magnetic materials and design. As a result, benchtop NMR has emerged as a powerful analytical tool for use in process analytical control (PAC). Nevertheless, the successful application of NMR devices as an analytical tool in several areas is intrinsically linked to its coupling with different chemometric methods. This review focuses on the evolution of benchtop NMR and chemometrics in chemical analysis, including applications in fuels, foods, pharmaceuticals, biochemicals, drugs, metabolomics, and polymers. The review also presents different low-resolution NMR methods for spectrum acquisition and chemometric techniques for calibration, classification, discrimination, data fusion, calibration transfer, multi-block and multi-way.
Collapse
Affiliation(s)
- Diego Galvan
- Chemistry Institute, Universidade Federal de Mato Grosso do Sul (UFMS), 79070-900, Campo Grande, MS, Brazil; Chemistry Departament, Universidade Estadual de Londrina (UEL), 86.057-970, Londrina, PR, Brazil.
| | | | - Evandro Bona
- Post-Graduation Program of Food Technology (PPGTA), Universidade Tecnológica Federal do Paraná (UTFPR), Campus Campo Mourão, 87301-899, Campo Mourão, PR, Brazil; Post-Graduation Program of Chemistry (PPGQ), Universidade Tecnológica Federal do Paraná (UTFPR), Campus Curitiba, 80230-901, Curitiba, PR, Brazil
| | - Federico Marini
- Department of Chemistry, University of Rome "La Sapienza", Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Mário Henrique M Killner
- Chemistry Departament, Universidade Estadual de Londrina (UEL), 86.057-970, Londrina, PR, Brazil
| |
Collapse
|
17
|
Matadh AV, Echanur A, Suresh S, Chede L, Maibach H, Kulkarni V, Murthy SN, H N S. Can Continuous Manufacturing of Topical Semisolids by Hot Melt Extrusion Soon Be a Reality? Mol Pharm 2023; 20:3779-3790. [PMID: 37421361 DOI: 10.1021/acs.molpharmaceut.3c00201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2023]
Abstract
For more than five decades, pharmaceutical manufacturers have been relying heavily on batch manufacturing that is a sequential, multistep, laborious, and time-consuming process. However, late advances in manufacturing technologies have prompted manufacturers to consider continuous manufacturing (CM) is a feasible manufacturing process that encompasses fewer steps and is less tedious and quick. Global regulatory agencies are taking a proactive role to facilitate pharmaceutical industries to adopt CM that assures product quality by employing robust manufacturing technologies encountering fewer interruptions, thereby substantially reducing product failures and recalls. However, adopting innovative CM is known to pose technical and regulatory challenges. Hot melt extrusion (HME) is one such state-of-the-art enabling technology that facilitates CM of diverse pharmaceutical dosage forms, including topical semisolids. Efforts have been made to continuously manufacture semisolids by HME integrating the principles of Quality by Design (QbD) and Quality Risk Management (QRM) and deploying Process Analytical Technologies (PAT) tools. Attempts have been made to systematically elucidate the effect of critical material attributes (CMA) and critical process parameters (CPP) on product critical quality attributes (CQA) and Quality Target Product Profiles (QTPP) deploying PAT tools. The article critically reviews the feasibility of one of the enabling technologies such as HME in CM of topical semisolids. The review highlights the benefits of the CM process and challenges ahead to implement the technology to topical semisolids. Once the CM of semisolids adopting melt extrusion integrated with PAT tools becomes a reality, the process can be extended to manufacture sterile semisolids that usually involve more critical processing steps.
Collapse
Affiliation(s)
- Anusha V Matadh
- Institute for Drug Delivery and Biomedical Research, Mahalaxmipuram, Bengaluru 560086, Karnataka, India
| | - Anusha Echanur
- Institute for Drug Delivery and Biomedical Research, Mahalaxmipuram, Bengaluru 560086, Karnataka, India
| | - Sarasija Suresh
- Institute for Drug Delivery and Biomedical Research, Mahalaxmipuram, Bengaluru 560086, Karnataka, India
| | - Laxmishanthi Chede
- College of Pharmacy, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Howard Maibach
- University of California, 2340 Sutter Street, San Francisco, California 94115, United States
| | - Vijay Kulkarni
- Steer Life Sciences, Fourth Phase, Peenya, Industrial Area, Bengaluru 560058, Karnataka, India
| | - S Narasimha Murthy
- Institute for Drug Delivery and Biomedical Research, Mahalaxmipuram, Bengaluru 560086, Karnataka, India
- Topical Products Testing, LLC, 9, Industrial Park Drive, Oxford, Mississippi 38655, United States
| | - Shivakumar H N
- Institute for Drug Delivery and Biomedical Research, Mahalaxmipuram, Bengaluru 560086, Karnataka, India
- KLE College of Pharmacy, Second Block, Rajajinagar, Bengaluru 560010, Karnataka, India
| |
Collapse
|
18
|
Optimal quantification of residence time distribution profiles from a quality assurance perspective. Int J Pharm 2023; 634:122653. [PMID: 36716830 DOI: 10.1016/j.ijpharm.2023.122653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/30/2023]
Abstract
Residence time distribution (RTD) has been widely applied across various fields of chemical engineering, including pharmaceutical manufacturing, for applications such as material traceability, quality assurance, system health monitoring, and fault detection. Determination of a representative RTD, in principle, requires an accurate process analytical technology (PAT) procedure capturing the entire range of tracer concentrations from zero to maximum. Such a wide concentration range creates at least two problems: i) decreased accuracy of the model across the entire range of concentrations, relating to limit of quantification, and ii) ambiguity associated with the detection of the tracer for low concentration levels, relating to limit of detection (LOD). These problems affect not only the RTD profile itself, but also RTD-based applications, which can potentially lead to erroneous conclusions. This article seeks to minimize the impact of these problems by understanding the relative importance of different features of RTD on the detection of out-of-specification (OOS) products. In this work, the RTD obtained experimentally was truncated at different levels, to investigate the impact of the truncation of RTD on funnel plots for OOS detection. The main finding is that the tail of the RTD can be truncated with no loss of accuracy in the determination of exclusion intervals. This enables the manufacturing scientist to focus entirely on the peak region, maximizing the accuracy of chemometric models.
Collapse
|
19
|
Jaspers M, Kulkarni SS, Tegel F, Roelofs TP, de Wit MT, Janssen PH, Meir B, Weinekötter R, Dickhoff BH. Batch versus continuous blending of binary and ternary pharmaceutical powder mixtures. Int J Pharm X 2022; 4:100111. [PMID: 35028558 PMCID: PMC8739470 DOI: 10.1016/j.ijpx.2021.100111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/15/2021] [Accepted: 12/30/2021] [Indexed: 12/03/2022] Open
Abstract
The material properties of excipients and active pharmaceutical ingredients (API's) are important parameters that affect blend uniformity of pharmaceutical powder formulations. With the current shift from batch to continuous manufacturing in the pharmaceutical industry, blending of excipients and API is converted to a continuous process. The relation between material properties and blend homogeneity, however, is generally based on batch-wise blending trials. Limited information is available on how material properties affect blending performance in a continuous process. Here, blending of API and excipients is studied in both a batch and a continuous process. Homogeneity of the resulting mixtures is analyzed, which reveals that the impact of material properties is very different in a continuous process. Where parameters such as particle size, density and flowability have significant impact on blending performance in a traditional batch process, continuous blending is more robust resulting in uniform blends for a large variety of blend compositions. Continuous mixing improves blend uniformity of pharmaceutical powder mixtures. Blend uniformity is highly dependent on excipient properties in a batch process. Continuous mixing is more robust, with little impact of material properties. Powder bulk density strongly affects blend homogeneity in a batch process. Blending of low API dosages is more challenging in a continuous process.
Collapse
|
20
|
Blue LE, Guan X, Joubert MK, Kuhns ST, Moore S, Semin DJ, Wikström M, Wypych J, Goudar CT. State-of-the-art and emerging trends in analytical approaches to pharmaceutical-product commercialization. Curr Opin Biotechnol 2022; 78:102800. [PMID: 36182871 DOI: 10.1016/j.copbio.2022.102800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 08/17/2022] [Accepted: 08/25/2022] [Indexed: 12/14/2022]
Abstract
The biopharmaceutical landscape continues to evolve rapidly, and associated modality complexity and the need to improve molecular understanding require concomitant advances in analytical approaches used to characterize and release the product. The Product Quality Attribute Assessment (PQAA) and Quality Target Product Profile (QTPP) frameworks help catalog and translate molecular understanding to process and product-design targets, thereby enabling reliable manufacturing of high-quality product. The analytical target profile forms the basis of identifying best-fit analytical methods for attribute measurement and continues to be successfully used to develop robust analytical methods for detailed product characterization as well as release and stability testing. Despite maturity across multiple testing platforms, advances continue to be made, several with the potential to alter testing paradigms. There is an increasing role for mass spectrometry beyond product characterization and into routine release testing as seen by the progress in multi-attribute methods and technologies, applications to aggregate measurement, the development of capillary zone electrophoresis (CZE) coupled with mass spectrometry (MS) and capillary isoelectric focusing (CIEF) with MS for measurement of glycans and charged species, respectively, and increased application to host cell protein measurement. Multitarget engaging multispecific modalities will drive advances in bioassay platforms and recent advances both in 1- and 2-D NMR approaches could make it the method of choice for characterizing higher-order structures. Additionally, rigorous understanding of raw material and container attributes is necessary to complement product understanding, and these collectively can enable robust supply of high-quality product to patients.
Collapse
Affiliation(s)
- Laura E Blue
- Attribute Sciences, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Xiaoyan Guan
- Attribute Sciences, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Marisa K Joubert
- Attribute Sciences, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Scott T Kuhns
- Attribute Sciences, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Stephanie Moore
- Attribute Sciences, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - David J Semin
- Attribute Sciences, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Mats Wikström
- Attribute Sciences, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Jette Wypych
- Attribute Sciences, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | - Chetan T Goudar
- Attribute Sciences, Process Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| |
Collapse
|
21
|
Parameter optimization in a continuous direct compression process of commercially batch-produced bisoprolol tablets. Int J Pharm 2022; 628:122355. [DOI: 10.1016/j.ijpharm.2022.122355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
|
22
|
Klinken S, Quodbach J. Sums of amplitudes analysis – A new non-parametric classification method for time series deviation evaluation in pharmaceutical processes. POWDER TECHNOL 2022. [DOI: 10.1016/j.powtec.2022.118003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
23
|
Be Rziņš KR, Mapley JI, Gordon KC, Fraser-Miller SJ. Evaluating Spatially Offset Low-Frequency Anti-Stokes Raman Spectroscopy (SOLFARS) for Detecting Subsurface Composition below an Emissive Layer: A Proof of Principle Study Using a Model Bilayer System. Mol Pharm 2022; 19:4311-4319. [PMID: 36170046 DOI: 10.1021/acs.molpharmaceut.2c00656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
This work explores the potential use of spatially offset low-frequency anti-Stokes Raman spectroscopy (SOLFARS) to detect subsurface composition below an emissive surface. A range of bilayer tablets were used to evaluate this approach. Bilayer tablets differed in both the underlying layer composition (active pharmaceutical ingredient to excipient ratio, celecoxib: α-lactose monohydrate) and the upper layer thickness of the fluorescent coating (polyvinylpyrrolidone mixture with sunset yellow FCF dye). Two low- (<300 cm-1) plus mid- (300 to 1800 cm-1) frequency Raman instrumental setups, with lateral displacements for spatial analysis of solid dosage forms, using different excitation wavelengths were explored. The 532 nm system was used to illustrate how the low-frequency anti-Stokes Raman approach works with samples exhibiting extreme fluorescence/background emission interference, and the 785 nm system was used to demonstrate the performance when less extreme fluorescence/emission is present. Qualitative and quantitative chemometric analyses were performed to evaluate the performance of individual spectral domains and their combinations for the determination of the composition of the subsurface layer as well as the coating layer thickness. Overall, the commonly used midfrequency region (300-1800 cm-1) proved superior when using 785 nm incident laser for quantifying the coating thickness (amorphous materials), whereas a combined Stokes and anti-Stokes low-frequency region was found to be superior for quantifying underlying crystalline materials. When exploring individual spectral regions for subsurface composition using spatially offset measurements, the anti-Stokes LFR spectral window performed best. The anti-Stokes low-frequency range also demonstrated an advantage for models composed of data exhibiting high levels of fluorescence (e.g., data collected using 532 nm incident laser), as the Stokes scattering was masked by fluorescence. Transmission measurements were also explored for comparison and showed the best applicability for both upper and lower layer analysis, attributed to the inherently larger bulk sampling volume of this setup. From a practical perspective, these results highlight the potential adjustments that can be made to already existing (in-line) Raman setups to facilitate similar analysis in pharmaceutical industry-based settings.
Collapse
Affiliation(s)
- Ka Rlis Be Rziņš
- The Dodd-Walls Centre for Photonic and Quantum Technologies, Department of Chemistry, University of Otago, Dunedin 9016, New Zealand.,Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 1165, Denmark
| | - Joseph I Mapley
- The Dodd-Walls Centre for Photonic and Quantum Technologies, Department of Chemistry, University of Otago, Dunedin 9016, New Zealand
| | - Keith C Gordon
- The Dodd-Walls Centre for Photonic and Quantum Technologies, Department of Chemistry, University of Otago, Dunedin 9016, New Zealand
| | - Sara J Fraser-Miller
- The Dodd-Walls Centre for Photonic and Quantum Technologies, Department of Chemistry, University of Otago, Dunedin 9016, New Zealand
| |
Collapse
|
24
|
Jelsch M, Roggo Y, Mohamad A, Kleinebudde P, Krumme M. Automatic system dynamics characterization of a pharmaceutical continuous production line. Eur J Pharm Biopharm 2022; 180:137-148. [PMID: 36122784 DOI: 10.1016/j.ejpb.2022.09.010] [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: 05/20/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/04/2022]
Abstract
Continuous Manufacturing (CM) of drug products is a new approach in the pharmaceutical industry. In the presented paper, a GMP continuous wet granulation line for production of solid oral dosage forms was investigated in order to assess the system dynamics of the line and to define the best control and diversion strategy. The following steps were involved in the continuous process: dosing / feeding, blending, twin-screw wet granulation, fluid-bed drying, sieving and tableting. Two drug products with two different drug substances were compared during this study: one drug substance as model drug compound and one formulation of a currently evaluated commercial drug product. Several step tests in API concentration were performed in order to characterize the process flow and assess the process dynamics. API content was monitored in real time by Process Analytical Technologies (PAT) thanks to three Near Infrared (NIR) probes located along the process and measuring the API content after blender, after dryer and in the tablet press feed frame. The process parameter values were changed during production in order to detect the impact on the quality of the final product. An automatic residence time distribution (RTD) computation method has been developed in order automate the RTD calculation on the basis of process data to further define and monitor the system dynamics with the final aim of out of specification material diversion during the continuous production. The RTD has been seen as a process fingerprint: a change in the RTD values implies a change in the process.
Collapse
Affiliation(s)
- Morgane Jelsch
- Novartis Pharma AG, Continuous Manufacturing (CM) Unit, CH-4002 Basel, Switzerland
| | - Yves Roggo
- Novartis Pharma AG, Continuous Manufacturing (CM) Unit, CH-4002 Basel, Switzerland
| | - Ahmad Mohamad
- Novartis Pharma AG, Continuous Manufacturing (CM) Unit, CH-4002 Basel, Switzerland
| | - Peter Kleinebudde
- Heinrich Heine University, Universitätsstraße1, 40225 Düsseldorf, Germany
| | - Markus Krumme
- Novartis Pharma AG, Continuous Manufacturing (CM) Unit, CH-4002 Basel, Switzerland.
| |
Collapse
|
25
|
Khanolkar A, Patil B, Thorat V, Samanta G. Development of Inline Near-Infrared Spectroscopy Method for Real-Time Monitoring of Blend Uniformity of Direct Compression and Granulation-Based Products at Commercial Scales. AAPS PharmSciTech 2022; 23:235. [PMID: 36002672 DOI: 10.1208/s12249-022-02392-9] [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: 06/28/2022] [Accepted: 08/08/2022] [Indexed: 11/30/2022] Open
Abstract
Blending is a critical intermediate unit operation for all solid oral formulations. For blend uniformity testing, API content in the blend must be quantified precisely. A detailed study was conducted to demonstrate the suitability of inline NIR (near-infrared) spectroscopy for blend uniformity testing of two solid oral formulations: existing direct compression (DC) product with a multistep blending process and granulation-based product with API granules. Both qualitative and quantitative methods were developed at a laboratory scale using statistical moving block standard deviation (MBSD) and multivariate data analysis such as principal component analysis (PCA) and partial least squares (PLS) regression. The qualitative MBSD method demonstrated that there was no need for multiple steps for the existing DC product. Hence, a simplified single-step process was developed for blending. Quantitative PLS models for blending processes of both the products were developed, validated, and successfully implemented at a commercial scale for the real-time release of blends. Results obtained from the validated model were in good agreement with the current method of sampling and chromatography.
Collapse
Affiliation(s)
- Aruna Khanolkar
- QbD Department, Integrated Product Development, Cipla Ltd., Maharashtra, Mumbai, India
| | - Bhaskar Patil
- QbD Department, Integrated Product Development, Cipla Ltd., Maharashtra, Mumbai, India
| | - Viraj Thorat
- QbD Department, Integrated Product Development, Cipla Ltd., Maharashtra, Mumbai, India
| | - Gautam Samanta
- QbD Department, Integrated Product Development, Cipla Ltd., Maharashtra, Mumbai, India.
| |
Collapse
|
26
|
Effect of batch-to-batch variation of spray dried lactose on the performance of feeders. POWDER TECHNOL 2022. [DOI: 10.1016/j.powtec.2022.117776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
27
|
Velez NL, Drennen JK, Anderson CA. Challenges, opportunities and recent advances in near infrared spectroscopy applications for monitoring blend uniformity in the continuous manufacturing of solid oral dosage forms. Int J Pharm 2022; 615:121462. [PMID: 35026317 DOI: 10.1016/j.ijpharm.2022.121462] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/20/2021] [Accepted: 01/07/2022] [Indexed: 10/19/2022]
Abstract
Near infrared (NIR) spectroscopy has been widely recognized as a powerful PAT tool for monitoring blend uniformity in continuous manufacturing (CM) processes. However, the dynamic nature of the powder stream and the fast rate at which it moves, compared to batch processes, introduces challenges to NIR quantitative methods for monitoring blend uniformity. For instance, defining the effective sample size interrogated by NIR, selecting the best sampling location for blend monitoring, and ensuring NIR model robustness against influential sources of variability are challenges commonly reported for NIR applications in CM. This article reviews the NIR applications for powder blend monitoring in the continuous manufacturing of solid oral dosage forms, with a particular focus on the challenges, opportunities for method optimization and recent advances with respect three main aspects: effective sample size measured by NIR, probe location and method robustness.
Collapse
Affiliation(s)
- Natasha L Velez
- Duquesne University Graduate School for Pharmaceutical Sciences, Pittsburgh, PA 15282, United States; Duquesne Center for Pharmaceutical Technology, Duquesne University, Pittsburgh, PA 15282, United States.
| | - James K Drennen
- Duquesne University Graduate School for Pharmaceutical Sciences, Pittsburgh, PA 15282, United States; Duquesne Center for Pharmaceutical Technology, Duquesne University, Pittsburgh, PA 15282, United States.
| | - Carl A Anderson
- Duquesne University Graduate School for Pharmaceutical Sciences, Pittsburgh, PA 15282, United States; Duquesne Center for Pharmaceutical Technology, Duquesne University, Pittsburgh, PA 15282, United States.
| |
Collapse
|
28
|
Sacher S, Poms J, Rehrl J, Khinast JG. PAT implementation for advanced process control in solid dosage manufacturing - A practical guide. Int J Pharm 2021; 613:121408. [PMID: 34952147 DOI: 10.1016/j.ijpharm.2021.121408] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/10/2021] [Accepted: 12/16/2021] [Indexed: 01/14/2023]
Abstract
The implementation of continuous pharmaceutical manufacturing requires advanced control strategies rather than traditional end product testing or an operation within a small range of controlled parameters. A high level of automation based on process models and hierarchical control concepts is desired. The relevant tools that have been developed and successfully tested in academic and industrial environments in recent years are now ready for utilization on the commercial scale. To date, the focus in Process Analytical Technology (PAT) has mainly been on achieving process understanding and quality control with the ultimate goal of real-time release testing (RTRT). This work describes the workflow for the development of an in-line monitoring strategy to support PAT-based real-time control actions and its integration into solid dosage manufacturing. All stages are discussed in this paper, from process analysis and definition of the monitoring task to technology assessment and selection, its process integration and the development of data acquisition.
Collapse
Affiliation(s)
- Stephan Sacher
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010 Graz, Austria.
| | - Johannes Poms
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010 Graz, Austria
| | - Jakob Rehrl
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010 Graz, Austria
| | - Johannes G Khinast
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010 Graz, Austria; Institute for Process and Particle Engineering, Graz University of Technology, Inffeldgasse 13/3, 8010 Graz, Austria
| |
Collapse
|
29
|
Bhalode P, Tian H, Gupta S, Razavi SM, Roman-Ospino A, Talebian S, Singh R, Scicolone JV, Muzzio FJ, Ierapetritou M. Using residence time distribution in pharmaceutical solid dose manufacturing - A critical review. Int J Pharm 2021; 610:121248. [PMID: 34748808 DOI: 10.1016/j.ijpharm.2021.121248] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/04/2021] [Accepted: 10/27/2021] [Indexed: 11/18/2022]
Abstract
While continuous manufacturing (CM) of pharmaceutical solid-based drug products has been shown to be advantageous for improving the product quality and process efficiency in alignment with FDA's support of the quality-by-design paradigm (Lee, 2015; Ierapetritou et al., 2016; Plumb, 2005; Schaber, 2011), it is critical to enable full utilization of CM technology for robust production and commercialization (Schaber, 2011; Byrn, 2015). To do so, an important prerequisite is to obtain a detailed understanding of overall process characteristics to develop cost-effective and accurate predictive models for unit operations and process flowsheets. These models are utilized to predict product quality and maintain desired manufacturing efficiency (Ierapetritou et al., 2016). Residence time distribution (RTD) has been a widely used tool to characterize the extent of mixing in pharmaceutical unit operations (Vanhoorne, 2020; Rogers and Ierapetritou, 2015; Teżyk et al., 2015) and manufacturing lines and develop computationally cheap predictive models. These models developed using RTD have been demonstrated to be crucial for various flowsheet applications (Kruisz, 2017; Martinetz, 2018; Tian, 2021). Though extensively used in the literature (Gao et al., 2012), the implementation, execution, evaluation, and assessment of RTD studies has not been standardized by regulatory agencies and can thus lead to ambiguity regarding their accurate implementation. To address this issue and subsequently prevent unforeseen errors in RTD implementation, the presented article aims to aid in developing standardized guidelines through a detailed review and critical discussion of RTD studies in the pharmaceutical manufacturing literature. The review article is divided into two main sections - 1) determination of RTD including different steps for RTD evaluation including experimental approach, data acquisition and pre-treatment, RTD modeling, and RTD metrics and, 2) applications of RTD for solid dose manufacturing. Critical considerations, pertaining to the limitations of RTDs for solid dose manufacturing, are also examined along with a perspective discussion of future avenues of improvement.
Collapse
Affiliation(s)
- Pooja Bhalode
- Department of Chemical and Biochemical Engineering, Rutgers - The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Huayu Tian
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | - Shashwat Gupta
- Department of Chemical and Biochemical Engineering, Rutgers - The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Sonia M Razavi
- Department of Chemical and Biochemical Engineering, Rutgers - The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Andres Roman-Ospino
- Department of Chemical and Biochemical Engineering, Rutgers - The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Shahrzad Talebian
- Department of Chemical and Biochemical Engineering, Rutgers - The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Ravendra Singh
- Department of Chemical and Biochemical Engineering, Rutgers - The State University of New Jersey, Piscataway, NJ 08854, USA
| | - James V Scicolone
- Department of Chemical and Biochemical Engineering, Rutgers - The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Fernando J Muzzio
- Department of Chemical and Biochemical Engineering, Rutgers - The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Marianthi Ierapetritou
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
30
|
Sacher S, Peter A, Khinast JG. Feasibility of In-line monitoring of critical coating quality attributes via OCT: Thickness, variability, film homogeneity and roughness. Int J Pharm X 2021; 3:100067. [PMID: 33385160 PMCID: PMC7772539 DOI: 10.1016/j.ijpx.2020.100067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 11/16/2022] Open
Abstract
The feasibility of Optical Coherence Tomography (OCT) for in-line monitoring of pharmaceutical film coating processes has recently been demonstrated. OCT enables real-time acquisition of high-resolution cross-sectional images of coating layers and computation of coating thickness. In addition, coating quality attributes can be computed based on in-line data. This study assesses the in-line applicability of OCT to various coating functionalities and formulations. Several types of commercial film-coated tablets containing the most common ingredients were investigated. To that end, the tablets were placed into a miniaturized perforated drum. An in-line OCT system was used to monitor the tablet bed. This set-up resembles the final stage of an industrial pan coating process. All investigated coatings were measured, and the coating thickness, homogeneity and roughness were computed. The rotation rate was varied in a range comparable to large-scale coating operations, and no influence on the outcome was observed. The results indicate that OCT can be used to determine end-point and establish in-process control for a wide range of coating formulations. The real-time computation of coating homogeneity and roughness can support process optimization and formulation development.
Collapse
Affiliation(s)
- Stephan Sacher
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010Graz, Austria
| | - Anna Peter
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010Graz, Austria
| | - Johannes G. Khinast
- Research Center Pharmaceutical Engineering GmbH, Inffeldgasse 13/2, 8010Graz, Austria
- Institute for Process and Particle Engineering, Graz University of Technology, Inffeldgasse 13/3, 8010 Graz, Austria
| |
Collapse
|
31
|
Towards real-time release of pharmaceutical tablets: 100% in-line control via near-infrared spatially resolved spectroscopy and 3D microwave resonance technology. J Pharm Biomed Anal 2021; 209:114491. [PMID: 34875571 DOI: 10.1016/j.jpba.2021.114491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 11/06/2021] [Accepted: 11/19/2021] [Indexed: 11/21/2022]
Abstract
In the scope of 100% in-line quality control and real-time release of pharmaceutical tablets, the authors present a flexible inspection module for in-line tablet analysis with integrated multipoint near-infrared (NIR) spectroscopy and 3D microwave resonance technology (3D MRT). Via an industrial case study on Diclofenac Sodium tablets, the abilities of this versatile process analytical technology (PAT) tool are presented. It is demonstrated that the combination of Diclofenac concentration prediction via NIR spectroscopy and mass prediction via 3D MRT allow to estimate the dosage of each individual tablet. Single sample repetition tests were performed on 5 tablets, measured 10 times on three different days. A high accuracy and precision of prediction was shown, with an average standard deviation below 0.5 mg. The inspection run demonstrated the added value of such inspection and sorting strategies based on the calculated dosage of individual tablets.
Collapse
|
32
|
Synthesis of a dipeptide by integrating a continuous flow reaction and continuous crystallization. Chem Eng Res Des 2021. [DOI: 10.1016/j.cherd.2021.09.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
33
|
Sohail Arshad M, Zafar S, Yousef B, Alyassin Y, Ali R, AlAsiri A, Chang MW, Ahmad Z, Ali Elkordy A, Faheem A, Pitt K. A review of emerging technologies enabling improved solid oral dosage form manufacturing and processing. Adv Drug Deliv Rev 2021; 178:113840. [PMID: 34147533 DOI: 10.1016/j.addr.2021.113840] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 12/20/2022]
Abstract
Tablets are the most widely utilized solid oral dosage forms because of the advantages of self-administration, stability, ease of handling, transportation, and good patient compliance. Over time, extensive advances have been made in tableting technology. This review aims to provide an insight about the advances in tablet excipients, manufacturing, analytical techniques and deployment of Quality by Design (QbD). Various excipients offering novel functionalities such as solubility enhancement, super-disintegration, taste masking and drug release modifications have been developed. Furthermore, co-processed multifunctional ready-to-use excipients, particularly for tablet dosage forms, have benefitted manufacturing with shorter processing times. Advances in granulation methods, including moist, thermal adhesion, steam, melt, freeze, foam, reverse wet and pneumatic dry granulation, have been proposed to improve product and process performance. Furthermore, methods for particle engineering including hot melt extrusion, extrusion-spheronization, injection molding, spray drying / congealing, co-precipitation and nanotechnology-based approaches have been employed to produce robust tablet formulations. A wide range of tableting technologies including rapidly disintegrating, matrix, tablet-in-tablet, tablet-in-capsule, multilayer tablets and multiparticulate systems have been developed to achieve customized formulation performance. In addition to conventional invasive characterization methods, novel techniques based on laser, tomography, fluorescence, spectroscopy and acoustic approaches have been developed to assess the physical-mechanical attributes of tablet formulations in a non- or minimally invasive manner. Conventional UV-Visible spectroscopy method has been improved (e.g. fiber-optic probes and UV imaging-based approaches) to efficiently record the dissolution profile of tablet formulations. Numerous modifications in tableting presses have also been made to aid machine product changeover, cleaning, and enhance efficiency and productivity. Various process analytical technologies have been employed to track the formulation properties and critical process parameters. These advances will contribute to a strategy for robust tablet dosage forms with excellent performance attributes.
Collapse
Affiliation(s)
| | - Saman Zafar
- Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Bushra Yousef
- Leicester School of Pharmacy, De Montfort University, Leicester, United Kingdom
| | - Yasmine Alyassin
- Leicester School of Pharmacy, De Montfort University, Leicester, United Kingdom
| | - Radeyah Ali
- Leicester School of Pharmacy, De Montfort University, Leicester, United Kingdom
| | - Ali AlAsiri
- Leicester School of Pharmacy, De Montfort University, Leicester, United Kingdom; Pharmacy College, Pharmaceutics Department, Najran University, Najran, Saudi Arabia
| | - Ming-Wei Chang
- Nanotechnology and Integrated Bioengineering Centre, University of Ulster, Jordanstown Campus, Newtownabbey BT37 0QB, Northern Ireland, United Kingdom
| | - Zeeshan Ahmad
- Leicester School of Pharmacy, De Montfort University, Leicester, United Kingdom
| | - Amal Ali Elkordy
- School of Pharmacy and Pharmaceutical Sciences, Faculty of Health Sciences and Wellbeing,University of Sunderland, Sunderland, United Kingdom
| | - Ahmed Faheem
- School of Pharmacy and Pharmaceutical Sciences, Faculty of Health Sciences and Wellbeing,University of Sunderland, Sunderland, United Kingdom; Faculty of Pharmacy, University of Tanta, Tanta, Egypt
| | - Kendal Pitt
- Manufacturing, Science & Technology, Pharma Supply Chain, GlaxoSmithKline, Ware, United Kingdom.
| |
Collapse
|
34
|
Domokos A, Pusztai É, Madarász L, Nagy B, Gyürkés M, Farkas A, Fülöp G, Casian T, Szilágyi B, Nagy ZK. Combination of PAT and mechanistic modeling tools in a fully continuous powder to granule line: Rapid and deep process understanding. POWDER TECHNOL 2021. [DOI: 10.1016/j.powtec.2021.04.059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
35
|
Abstract
Spray drying is a versatile technology that has been applied widely in the chemical, food, and, most recently, pharmaceutical industries. This review focuses on engineering advances and the most significant applications of spray drying for pharmaceuticals. An in-depth view of the process and its use is provided for amorphous solid dispersions, a major, growing drug-delivery approach. Enhanced understanding of the relationship of spray-drying process parameters to final product quality attributes has made robust product development possible to address a wide range of pharmaceutical problem statements. Formulation and process optimization have leveraged the knowledge gained as the technology has matured, enabling improved process development from early feasibility screening through commercial applications. Spray drying's use for approved small-molecule oral products is highlighted, as are emerging applications specific to delivery of biologics and non-oral delivery of dry powders. Based on the changing landscape of the industry, significant future opportunities exist for pharmaceutical spray drying.
Collapse
Affiliation(s)
- John M Baumann
- Small Molecules, Lonza Pharma & Biotech, Bend, Oregon 97701, USA; , ,
| | - Molly S Adam
- Small Molecules, Lonza Pharma & Biotech, Bend, Oregon 97701, USA; , ,
| | - Joel D Wood
- Small Molecules, Lonza Pharma & Biotech, Bend, Oregon 97701, USA; , ,
| |
Collapse
|
36
|
Hu C. Reactor design and selection for effective continuous manufacturing of pharmaceuticals. J Flow Chem 2021; 11:243-263. [PMID: 34026279 PMCID: PMC8130218 DOI: 10.1007/s41981-021-00164-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/14/2021] [Indexed: 11/23/2022]
Abstract
Pharmaceutical production remains one of the last industries that predominantly uses batch processes, which are inefficient and can cause drug shortages due to the long lead times or quality defects. Consequently, pharmaceutical companies are transitioning away from outdated batch lines, in large part motivated by the many advantages of continuous manufacturing (e.g., low cost, quality assurance, shortened lead time). As chemical reactions are fundamental to any drug production process, the selection of reactor and its design are critical to enhanced performance such as improved selectivity and yield. In this article, relevant theories, and models, as well as their required input data are summarized to assist the reader in these tasks, focusing on continuous reactions. Selected examples that describe the application of plug flow reactors (PFRs) and continuous-stirred tank reactors (CSTRs)-in-series within the pharmaceutical industry are provided. Process analytical technologies (PATs), which are important tools that provide real-time in-line continuous monitoring of reactions, are recommended to be considered during the reactor design process (e.g., port design for the PAT probe). Finally, other important points, such as density change caused by thermal expansion or solid precipitation, clogging/fouling, and scaling-up, are discussed. Graphical abstract
Collapse
Affiliation(s)
- Chuntian Hu
- CONTINUUS Pharmaceuticals, Woburn, MA 01801 USA
| |
Collapse
|
37
|
Alam MA, Liu YA, Dolph S, Pawliczek M, Peeters E, Palm A. Benchtop NIR method development for continuous manufacturing scale to enable efficient PAT application for solid oral dosage form. Int J Pharm 2021; 601:120581. [PMID: 33839228 DOI: 10.1016/j.ijpharm.2021.120581] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/24/2021] [Accepted: 04/04/2021] [Indexed: 11/15/2022]
Abstract
A Near Infrared (NIR) method was developed using a small benchtop feed frame system to quantify Saccharin potency in a powder blend during continuous manufacturing process. A 15-point Design of Experiments (DoE) was created based on the NIR spectral response and compositions of the formulation to develop a calibration set. The calibration set was designed to create compositional and raw material lots variation using minimum resources. The calibration experiments utilized around 0.5 kg Saccharin (Active Pharmaceutical Ingredient (API) surrogate) and 1.8 kg of excipients. Partial Least Square (PLS) modeling was used to develop a quantitative NIR method from the calibration data. The NIR method was implemented during 5 test batches in two different manufacturing sites across different potency levels at a continuous manufacturing platform for direction compression. Acceptable prediction performance was achieved from the NIR method at both sites. The NIR method was robust against changes in process scale and NIR instruments. The variance information built into the calibration set was found to be critical to successful model performance. This study shows a benchtop feed frame can be used for material sparing calibration method development without operating at a full-scale process line and applied across multiple sites, instruments at different potency levels.
Collapse
Affiliation(s)
- Md Anik Alam
- Worldwide Research and Development, Pfizer Inc., Groton, CT 06340, USA.
| | - Yang Angela Liu
- Worldwide Research and Development, Pfizer Inc., Groton, CT 06340, USA
| | - Stephanie Dolph
- Worldwide Research and Development, Pfizer Inc., Groton, CT 06340, USA
| | - Marcin Pawliczek
- Pfizer Global Supply, Pfizer Manufacturing Deutschland GmbH, Freiburg, Germany
| | - Elisabeth Peeters
- Worldwide Research and Development, Pfizer Inc., Groton, CT 06340, USA
| | - Andrew Palm
- Worldwide Research and Development, Pfizer Inc., Groton, CT 06340, USA
| |
Collapse
|
38
|
Jaspers M, de Wit MT, Kulkarni SS, Meir B, Janssen PH, van Haandel MM, Dickhoff BH. Impact of excipients on batch and continuous powder blending. POWDER TECHNOL 2021. [DOI: 10.1016/j.powtec.2021.02.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
39
|
Higgins H, Roeing-Donna M, Krupiarz K, O'Connor R, Tan J, Pohlman NA. Granular flow behavior in a conveyor system: From local velocity profiles to mass flow rates. POWDER TECHNOL 2021. [DOI: 10.1016/j.powtec.2020.12.062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
40
|
Igne B, Liu Y, Shi Z, Alam MA, Garrett A, Daughtry S, Liesum L, Nielsen S. Multivariate Spectroscopic Method Lifecycle Management as Part of the Quality Management System. J Pharm Sci 2021; 110:2925-2933. [PMID: 33785351 DOI: 10.1016/j.xphs.2021.03.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 10/21/2022]
Abstract
Multivariate model based spectroscopic methods require model maintenance through their lifecycle. A survey conducted by the International Consortium for Innovation and Quality in Pharmaceutical Development (IQ) in 2019 showed that regulatory reporting categories for the model related changes can be a hurdle for the routine use of these types of methods. This article introduces industry best practices on multivariate method and model lifecycle management within the Pharmaceutical Quality System. Case studies are provided to demonstrate how the Established Conditions and Post-Approval Change Management Protocol concepts may be leveraged to allow regulatory flexibility for change management and to encourage the use of these techniques for the development and commercialization of pharmaceutical products.
Collapse
Affiliation(s)
- Benoît Igne
- Analytical Development, Vertex Pharmaceuticals Inc., Boston, MA, USA.
| | - Yang Liu
- Pfizer, Worldwide Research and Development, Analytical R&D, Groton, CT, USA
| | - Zhenqi Shi
- Lilly Research Laboratory, Eli Lilly and Company, Indianapolis, IN, USA
| | - Md Anik Alam
- Pfizer, Worldwide Research and Development, Analytical R&D, Groton, CT, USA
| | - Aaron Garrett
- Global Quality Laboratory, Eli Lilly and Company, Indianapolis, IN, USA
| | - Sean Daughtry
- Analytical Development, Vertex Pharmaceuticals Inc., Boston, MA, USA
| | - Lorenz Liesum
- Roche, Pharma Technical Innovation, F. Hoffmann- La Roche Ltd, 4070 Basel, Switzerland
| | - Sarah Nielsen
- Janssen Supply Chain, Johnson & Johnson, New Brunswick, NJ, USA
| |
Collapse
|
41
|
Domokos A, Nagy B, Szilágyi B, Marosi G, Nagy ZK. Integrated Continuous Pharmaceutical Technologies—A Review. Org Process Res Dev 2021. [DOI: 10.1021/acs.oprd.0c00504] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- András Domokos
- Budapest University of Technology and Economics, Organic Chemistry and Technology Department, H-1111 Budapest, Hungary
| | - Brigitta Nagy
- Budapest University of Technology and Economics, Organic Chemistry and Technology Department, H-1111 Budapest, Hungary
| | - Botond Szilágyi
- Budapest University of Technology and Economics, Faculty of Chemical Technology and Biotechnology, H-1111 Budapest, Hungary
| | - György Marosi
- Budapest University of Technology and Economics, Organic Chemistry and Technology Department, H-1111 Budapest, Hungary
| | - Zsombor Kristóf Nagy
- Budapest University of Technology and Economics, Organic Chemistry and Technology Department, H-1111 Budapest, Hungary
| |
Collapse
|
42
|
Testa CJ, Shvedova K, Hu C, Wu W, Born SC, Takizawa B, Mascia S. Heterogeneous Crystallization as a Process Intensification Technology in an Integrated Continuous Manufacturing Process for Pharmaceuticals. Org Process Res Dev 2021. [DOI: 10.1021/acs.oprd.0c00468] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Christopher J. Testa
- CONTINUUS Pharmaceuticals, 25R Olympia Avenue, Woburn, Massachusetts 01801, United States
| | - Khrystyna Shvedova
- CONTINUUS Pharmaceuticals, 25R Olympia Avenue, Woburn, Massachusetts 01801, United States
| | - Chuntian Hu
- CONTINUUS Pharmaceuticals, 25R Olympia Avenue, Woburn, Massachusetts 01801, United States
| | - Wei Wu
- CONTINUUS Pharmaceuticals, 25R Olympia Avenue, Woburn, Massachusetts 01801, United States
| | - Stephen C. Born
- CONTINUUS Pharmaceuticals, 25R Olympia Avenue, Woburn, Massachusetts 01801, United States
| | - Bayan Takizawa
- CONTINUUS Pharmaceuticals, 25R Olympia Avenue, Woburn, Massachusetts 01801, United States
| | - Salvatore Mascia
- CONTINUUS Pharmaceuticals, 25R Olympia Avenue, Woburn, Massachusetts 01801, United States
| |
Collapse
|
43
|
El‐Sayed GM, El Mously DA, Mostafa NM, Hassan NY, Mahmoud AM. Design of Copper Microfabricated Potentiometric Sensor for
in‐line
Monitoring of Neostigmine Degradation Kinetics. ELECTROANAL 2021. [DOI: 10.1002/elan.202060536] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Ghada M. El‐Sayed
- Analytical Chemistry Department Faculty of Pharmacy Cairo University Kasr-El-Aini 11562 Cairo Egypt
| | - Dina A. El Mously
- Analytical Chemistry Department Faculty of Pharmacy Cairo University Kasr-El-Aini 11562 Cairo Egypt
| | - Nadia M. Mostafa
- Analytical Chemistry Department Faculty of Pharmacy Cairo University Kasr-El-Aini 11562 Cairo Egypt
| | - Nagiba Y. Hassan
- Analytical Chemistry Department Faculty of Pharmacy Cairo University Kasr-El-Aini 11562 Cairo Egypt
| | - Amr M. Mahmoud
- Analytical Chemistry Department Faculty of Pharmacy Cairo University Kasr-El-Aini 11562 Cairo Egypt
| |
Collapse
|
44
|
Köster C, Pohl S, Kleinebudde P. Evaluation of Binders in Twin-Screw Wet Granulation. Pharmaceutics 2021; 13:pharmaceutics13020241. [PMID: 33572394 PMCID: PMC7916237 DOI: 10.3390/pharmaceutics13020241] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 11/16/2022] Open
Abstract
The binders povidone (Kollidon 30), copovidone (Kollidon VA64), hypromellose (Pharmacoat 606), and three types of hyprolose (HPC SSL‑SFP, HPC SSL, and HPC SL‑FP) were evaluated regarding their suitability in twin-screw wet granulation. Six mixtures of lactose and binder as well as lactose without binder were twin-screw granulated with demineralized water at different barrel fill levels and subsequently tableted. A screening run with HPC SSL determined the amount of water as an influential parameter for oversized agglomerates. Subsequent examination of different binders, especially Kollidon 30 and Kollidon VA64 resulted in large granules. All binders, except Pharmacoat 606, led to a reduction of fines compared to granulation without a binder. The molecular weight of applied hyproloses did not appear as influential. Tableting required an upstream sieving step to remove overlarge granules. Tableting was possible for all formulations at sufficient compression pressure. Most binders resulted in comparable tensile strengths, while Pharmacoat 606 led to lower and lactose without a binder to the lowest tensile strength. Tablets without a binder disintegrated easily, whereas binder containing tablets of sufficient tensile strength often nearly failed or failed the disintegration test. Especially tablets containing Pharmacoat 606 and HPC SL‑FP disintegrated too slowly.
Collapse
|
45
|
Narala S, Nyavanandi D, Srinivasan P, Mandati P, Bandari S, Repka MA. Pharmaceutical Co-Crystals, Salts, and Co-Amorphous Systems: A Novel Opportunity of Hot Melt Extrusion. J Drug Deliv Sci Technol 2021; 61:102209. [PMID: 33717230 PMCID: PMC7946067 DOI: 10.1016/j.jddst.2020.102209] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Enhancing the solubility of active drug ingredients is a major challenge faced by scientists and researchers. Different approaches have been explored for the enhancement of solubility and physicochemical properties of drugs, without affecting their stability or pharmacological activity. Among the various strategies available, pharmaceutical co-crystals, co-amorphous systems, and pharmaceutical salts as multicomponent systems (MCS) have gained interest to improve physicochemical properties of drugs. Development of MCS by conventional methods involves the utilization of excess amount of solvents, thus, making the product prone to instability, and may also cause harmful side effects in patients. Scale up is critical and involves the investment of huge capital and time. Lately, hot-melt extrusion has been utilized in the development of MCS to enhance solubility, bioavailability, stability, and physicochemical properties of the drugs. In this review, the authors discussed the development of different MCS produced via hot-melt extrusion technology. Specifically, approaches for screening of co-formers and co-crystals, selection of excipients for co-amorphous systems, pharmaceutical salts, and significance of MCS and process parameters affecting product quality are discussed.
Collapse
Affiliation(s)
- Sagar Narala
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38677, USA
| | - Dinesh Nyavanandi
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38677, USA
| | - Priyanka Srinivasan
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38677, USA
| | - Preethi Mandati
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38677, USA
| | - Suresh Bandari
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38677, USA
| | - Michael A. Repka
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38677, USA
- Pii Center for Pharmaceutical Technology, The University of Mississippi, University, MS 38677, USA
| |
Collapse
|
46
|
Abstract
Tableting by direct compression (DC) is one of the simplest and most cost-effective drug manufacturing approaches. However, most active pharmaceutical ingredients (APIs) and excipients lack the compression and flow properties required to meet the needs of high-speed industrial tablet presses. Therefore, the majority of DC APIs and excipients are modified via processing/co-processing particle engineering techniques to boost their properties. Spray drying is one of the most commonly employed techniques to prepare DC grades of APIs and excipients with prominent advantages. This review aims to present an overview of the commercially marketed and investigationally-prepared DC APIs and excipients produced by spray drying.
Collapse
|
47
|
Shibayama S, Funatsu K. Investigation of Preprocessing and Validation Methodologies for PAT: Case Study of the Granulation and Coating Steps for the Manufacturing of Ethenzamide Tablets. AAPS PharmSciTech 2021; 22:41. [PMID: 33420526 DOI: 10.1208/s12249-020-01911-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 12/18/2020] [Indexed: 11/30/2022] Open
Abstract
After the Food and Drug Association in the USA published guidelines on the enhanced use of process analytical technology (PAT) and continuous manufacturing, many studies regarding PAT and continuous manufacturing have been published. This paper describes a case study involving granulation and coating steps with ethenzamide to investigate interference for PAT model construction and model management. We investigated what factors should be considered and addressed when PAT is implemented for continuous manufacturing and how predictive models should be constructed. The product qualities that were monitored were moisture content and particle size in the granulation step and tablet weight and moisture content in the coating step. We have constructed models for the granulation step and validated the predictive capability of the models against an external dataset. A partial least squares (PLS) model with manual wavelength selection had the best predictive accuracy for loss on drying against the external validation set. We found that the prediction of loss on drying was accurate, but the prediction of particle size was not sufficiently accurate. In the coating step, because of the small amount of data, we performed three-fold cross-validation and y-scrambling 10 times, to select the optimal hyper-parameters and to check if the models were fitted to chance correlations. We confirmed that the coating agent weights, tablet weights, and water content could be accurately predicted based on the mean of the R2 score for cross-validation. Addition of other variables, as well as the absorbance, slightly improved the predictive accuracy.
Collapse
|
48
|
Implementing Feedback Granule Size Control in a Continuous Dry Granulation Line Using Controlled Impeller Speed of the Granulation Unit, Compaction Force and Gap Width. J Pharm Innov 2020. [DOI: 10.1007/s12247-020-09524-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Abstract
Purpose
In continuous manufacturing of pharmaceuticals, dry granulation is of interest because of its large throughput capacity and energy efficiency. In order to manufacture solid oral dosage forms continuously, valid control strategies for critical quality attributes should be established. To this date, there are no published control strategies for granule size distribution in continuous dry granulation.
Methods
In-line laser diffraction was used to determine the size of granules in a continuous roll compaction/dry granulation line (QbCon® dry). Different process parameters were evaluated regarding their influences on granule size. The identified critical process parameters were then incorporated into control strategies. The uncontrolled and the controlled processes were compared based on the resulting granule size. In both processes, a process parameter was changed to induce a shift in median particle size and the controller had to counteract this shift.
Results
In principle, all process parameters that affect the median particle size could also be used to control the particle size in a dry granulation process. The sieve impeller speed was found to be well suited to control the median particle size as it reacts fast and can be controlled independently of the throughput or material.
Conclusion
The median particle size in continuous roll compaction can be controlled by adjusting process parameters depending on real-time granule size measurements. The method has to be validated and explored further to identify critical requirements to the material and environmental conditions.
Collapse
|
49
|
Vo AQ, Kutz G, He H, Narala S, Bandari S, Repka MA. Continuous Manufacturing of Ketoprofen Delayed Release Pellets Using Melt Extrusion Technology: Application of QbD Design Space, Inline Near Infrared, and Inline Pellet Size Analysis. J Pharm Sci 2020; 109:3598-3607. [PMID: 32916139 PMCID: PMC7680423 DOI: 10.1016/j.xphs.2020.09.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/27/2020] [Accepted: 09/03/2020] [Indexed: 11/27/2022]
Abstract
Delayed-release dosage forms are mainly manufactured as batch processes and include coated tablets, pellets, or particles with gastric resistant polymers. Authors propose a novel approach using the hot-melt extrusion technique to prepare delayed release dosage forms via a continuous manufacturing process, a new trend in the pharmaceutical industry. A full factorial design was employed to correlate input variables, including stearic acid (SA) content, drug content, and pellet size with drug release properties of the pellets. PLS fit method suitably elaborated the relationship between input and output variables with reasonably good fit and goodness of prediction. All three input factors influenced drug release in enzyme-free simulated gastric fluid (SGF) after 120 min; however, SA content did not significantly affect drug dissolution in the enzyme-free simulated intestinal fluid (SIF). An optimized formulation and design space were determined by overlaying multiple contours established from regression equations. The continuous manufacturing process was successfully monitored using inline near-infrared (NIR) and inline particle size analysis, with drug load and pellet size being well-controlled within the design space. The obtained pellets released less than 5% after 120 min in SGF and more than 85% and 95% after 30 min and 45 min, respectively, after switching to SIF.
Collapse
Affiliation(s)
- Anh Q Vo
- School of Pharmacy, University of Mississippi, University, MS 38677, USA; Department of Physical Chemistry and Physics, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Gerd Kutz
- OWL University of Applied Sciences and Arts, Pharmaceutical Engineering, Lemgo, Germany
| | - Herman He
- Thermo Fisher Scientific, Tewksbury, MA 01876, USA
| | - Sagar Narala
- School of Pharmacy, University of Mississippi, University, MS 38677, USA
| | - Suresh Bandari
- School of Pharmacy, University of Mississippi, University, MS 38677, USA
| | - Michael A Repka
- School of Pharmacy, University of Mississippi, University, MS 38677, USA; Pii Center for Pharmaceutical Technology, The University of Mississippi, University, MS 38677, USA.
| |
Collapse
|
50
|
UPLC-PDA: A greener miniaturized tool for the analysis and purity assessment of ebastine and phenylephrine hydrochloride. Microchem J 2020. [DOI: 10.1016/j.microc.2020.105400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|