1
|
Mazdi NTA, Mior Mat Zin NA, Khairul Hisham MA, Mohd Rus S, Haris MS, Chatterjee B. Optimizing Paracetamol-Ascorbic Acid Effervescent Tablet Characteristics: A Quality by Design Approach . Drug Dev Ind Pharm 2025:1-19. [PMID: 40253623 DOI: 10.1080/03639045.2025.2495131] [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: 10/18/2024] [Revised: 04/04/2025] [Accepted: 04/13/2025] [Indexed: 04/22/2025]
Abstract
OBJECTIVE This study aims to optimize paracetamol-ascorbic acid (PCM-AA) effervescent tablet characteristics through a Quality-by-Design (QbD) approach, investigating the effects of binder concentration, granulation time, and effervescent agents' ratio on hardness, disintegration, and dissolution. METHODS QbD approach was implemented by identifying the quality target product profile, critical quality attributes (CQAs), critical material attributes (CMAs), and critical process parameters for formulating PCM-AA effervescent tablets. An Ishikawa diagram identified risk factors for CQAs. A risk estimation matrix evaluated the levels of associated risks. A central composite design-based response surface methodology with 20 experimental runs, including six center points, identified key factors (binder concentration, granulation time, and effervescent agents' ratio) influencing tablet characteristics (hardness, disintegration, dissolution). The optimum formulation, determined by numerical analysis, was characterized for weight uniformity, tablet thickness and diameter, friability, and PCM and AA assay. RESULTS Optimized PCM (500 mg)-AA(200mg) effervescent tablets with 2.9% PVP concentration, 15 min granulation time, and 1:1.5 sodium bicarbonate-citric acid ratio achieved acceptable characteristics (hardness: 45N ± 20N, disintegration: <5 min, and both PCM and AA dissolution: <10 min). Model validation showed no significant difference (p > 0.05), indicating consistent results. CONCLUSION The study successfully optimized the hardness, disintegration, and dissolution rate of PCM-AA effervescent tablets via the QbD approach. Granulation time affects hardness and PCM dissolution, binder concentration influences disintegration time, and the effervescent agents' ratio impacts both disintegration time and AA dissolution. This research enhances the understanding of pharmaceutical formulation processes, risk management, and optimization in effervescent tablet development.
Collapse
Affiliation(s)
- Nur Tasnim Adlina Mazdi
- Department of Pharmaceutical Technology, Kulliyyah of Pharmacy, International Islamic University Malaysia, 25200 Kuantan, Pahang, Malaysia
| | - Nur Aisyah Mior Mat Zin
- Department of Pharmaceutical Technology, Kulliyyah of Pharmacy, International Islamic University Malaysia, 25200 Kuantan, Pahang, Malaysia
| | - Muhammad Aiman Khairul Hisham
- Department of Pharmaceutical Technology, Kulliyyah of Pharmacy, International Islamic University Malaysia, 25200 Kuantan, Pahang, Malaysia
| | - Shaiqah Mohd Rus
- Department of Pharmaceutical Technology, Universiti Kuala Lumpur Royal College of Medicine Perak, 30450 Ipoh, Perak, Malaysia
| | - Muhammad Salahuddin Haris
- Department of Pharmaceutical Technology, Kulliyyah of Pharmacy, International Islamic University Malaysia, 25200 Kuantan, Pahang, Malaysia
- Department of Pharmaceutical Technology, Universiti Kuala Lumpur Royal College of Medicine Perak, 30450 Ipoh, Perak, Malaysia
| | | |
Collapse
|
2
|
Beg S, Ahirwar K, Almalki WH, Almujri SS, Alhamyani A, Rahman M, Shukla R. Nondestructive techniques for pharmaceutical drug product characterization. Drug Discov Today 2025; 30:104249. [PMID: 39580022 DOI: 10.1016/j.drudis.2024.104249] [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: 03/31/2024] [Revised: 09/22/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024]
Abstract
Pharmaceutical product development involves multiple steps; therefore product quality must be assessed to ensure robustness and acceptability. Raw components, production methods, and ambient conditions yield highly variable end products with low batch-to-batch consistency. Although end testing is performed to ensure product quality, intermediate quality checks are limited. Nondestructive techniques like terahertz, near-infrared, X-ray, and Raman spectroscopy are common tools for in-line quality checks and real-time data monitoring. Handheld devices based on these analytical techniques also help in identifying counterfeit drugs products. This review discusses modern regulatory perspectives on the use of nondestructive tools in pharmaceutical quality monitoring.
Collapse
Affiliation(s)
- Sarwar Beg
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Kailash Ahirwar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Lucknow 226002, India
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Salem S Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Asir-Abha 61421, Saudi Arabia
| | - Abdulrahman Alhamyani
- Pharmaceuticals Chemistry Department, Faculty of Clinical Pharmacy, Al Baha University, Al Baha 65779, Saudi Arabia
| | - Mahfoozur Rahman
- Department of Pharmaceutical Sciences, Shalom Institute of Health & Allied Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Lucknow 226002, India.
| |
Collapse
|
3
|
Muñoz López C, Peeters K, Van Impe J. Data-Driven Modeling of the Spray Drying Process. Process Monitoring and Prediction of the Particle Size in Pharmaceutical Production. ACS OMEGA 2024; 9:25678-25693. [PMID: 38911742 PMCID: PMC11191099 DOI: 10.1021/acsomega.3c08032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 02/07/2024] [Accepted: 03/05/2024] [Indexed: 06/25/2024]
Abstract
Spray drying is used in the pharmaceutical industry for particle engineering of amorphous solid dispersions (ASDs). The particle size of the spray-dried (SD) powders is one of their key attributes due to its impact on the downstream processes and the drug product's functional properties. Offline and inline laser diffraction methods can be used to estimate the product's particle size; however, the final release of these ASDs is based on offline instruments. This paper presents a novel data-driven modeling approach for predicting the particle size of SD products. The model-based characterization of the process and the product's particle size, as a critical quality attribute, follows the quality by design principles. The resulting model can be used for online process monitoring, reducing the risks of out-of-specifications products and supporting their real-time release. A Tucker3 model is trained to capture and factorize the deterministic variability of the process. Subsequently, a partial least-squares regression model is calibrated to model the impact that variability in the input material properties, the process parameters, and the spray nozzle have on the products' particle size. This strategy has been calibrated and validated using large scale production data for two intermediate drug products under high sparsity of particle size data. Despite the challenges, high accuracy was obtained in predicting the median particle size (dv50) for release. The 99% confidence interval results in an error of maximum 2.5 μm, which is less than 10% of the allowed range of variation.
Collapse
Affiliation(s)
- Carlos
André Muñoz López
- BioTeC+
Chemical & Biochemical Process Technology & Control, Campus
Gent, KU Leuven, Gebroeders De Smetstraat 1, 9000 Ghent, Belgium
| | - Kristin Peeters
- Technical
Operations, Geel Chemical Production Site, Janssen Pharmaceutica, J&J, Janssen-Pharmaceuticalaan 3, 2440 Geel, Belgium
| | - Jan Van Impe
- BioTeC+
Chemical & Biochemical Process Technology & Control, Campus
Gent, KU Leuven, Gebroeders De Smetstraat 1, 9000 Ghent, Belgium
| |
Collapse
|
4
|
Simão J, Chaudhary SA, Ribeiro AJ. Implementation of Quality by Design (QbD) for development of bilayer tablets. Eur J Pharm Sci 2023; 184:106412. [PMID: 36828037 DOI: 10.1016/j.ejps.2023.106412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/20/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023]
Abstract
Bilayer tablets offer various drug release profiles for individual drugs incorporated in each layer of a bilayer tablet, which is rarely achievable by conventional tablets. These tablets also help avoid physicochemical incompatibilities between drugs and excipients. Successful manufacturing of such more complex dosage forms depends upon screening of material attributes of API and excipients as well as optimization of processing parameters of individual unit operations of the manufacturing process that must be strictly monitored and controlled to obtain an acceptable drug product quality and performance in order to achieve safety and efficacy per regulatory requirements. Optimizing formulation attributes and manufacturing processes during critical stages, such as blending, granulation, pre-compression, and main compression, can help avoid problems such as weight variation, segregation, and delamination of individual layers, which are frequently faced during the production of bilayer tablets. The main objective of this review is to establish the basis for the implementation of Quality by Design (QbD) system principles for the design and development of bilayer tablets, encompassing the preliminary and systematic risk assessment of critical material attributes (CMAs) and critical process parameters (CPPs) with respect to in-process and finished product critical quality attributes (CQAs). Moreover, the applicability of the QbD methodology based on its purpose is discussed and complemented with examples of bilayer tablet technology.
Collapse
Affiliation(s)
- J Simão
- Faculdade de Farmácia, Universidade de Coimbra, Coimbra, Portugal
| | - S A Chaudhary
- National Institute of Pharmaceutical Education and Research, Ahmedabad, India
| | - A J Ribeiro
- Faculdade de Farmácia, Universidade de Coimbra, Coimbra, Portugal; i3S, IBMC, Rua Alfredo Allen, Porto, Portugal.
| |
Collapse
|
5
|
Development and Evaluation of Cannabidiol Orodispersible Tablets Using a 23-Factorial Design. Pharmaceutics 2022; 14:pharmaceutics14071467. [PMID: 35890362 PMCID: PMC9324952 DOI: 10.3390/pharmaceutics14071467] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 01/23/2023] Open
Abstract
Orodispersible tablets (ODTs) are pharmaceutical formulations used to obtain fast therapeutic effects, usually recommended for geriatric and pediatric patients due to their improved compliance, bioavailability, ease of administration, and good palatability. This study aimed to develop ODTs with cannabidiol (CBD) phytocannabinoid extracted from Cannabis sativa used in the treatment of Lennox–Gastaut and Dravet syndromes. The tablets were obtained using an eccentric tableting machine and 9 mm punches. To develop CBD ODTs, the following parameters were varied: the Poloxamer 407 concentration (0 and 10%), the type of co-processed excipient (Prosolv® ODT G2—PODTG2 and Prosolv® EasyTab sp—PETsp), and the type of superdisintegrant (Croscarmellose—CCS, and Soy Polysaccharides—Emcosoy®—EMCS), resulting in eleven formulations (O1–O11). The following dependent parameters were evaluated: friability, disintegration time, crushing strength, and the CBD dissolution at 1, 3, 5, 10, 15, and 30 min. The dependent parameters were verified according to European Pharmacopoeia (Ph. Eur.) requirements. All the tablets obtained were in accordance with quality requirements in terms of friability (less than 1%), and disintegration time (less than 180 s). The crushing strength was between 19 N and 80 N. Regarding the dissolution test, only four formulations exhibited an amount of CBD released higher than 80% at 30 min. Taking into consideration the results obtained and using the Modde 13.1 software, an optimal formulation was developed (O12), which respected the quality criteria chosen (friability 0.23%, crushing strength of 37 N, a disintegration time of 27 s, and the target amount of CBD released in 30 min of 99.3 ± 6%).
Collapse
|
6
|
Kim EJ, Kim JH, Kim MS, Jeong SH, Choi DH. Process Analytical Technology Tools for Monitoring Pharmaceutical Unit Operations: A Control Strategy for Continuous Process Verification. Pharmaceutics 2021; 13:919. [PMID: 34205797 PMCID: PMC8234957 DOI: 10.3390/pharmaceutics13060919] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/31/2021] [Accepted: 06/16/2021] [Indexed: 11/16/2022] Open
Abstract
Various frameworks and methods, such as quality by design (QbD), real time release test (RTRT), and continuous process verification (CPV), have been introduced to improve drug product quality in the pharmaceutical industry. The methods recognize that an appropriate combination of process controls and predefined material attributes and intermediate quality attributes (IQAs) during processing may provide greater assurance of product quality than end-product testing. The efficient analysis method to monitor the relationship between process and quality should be used. Process analytical technology (PAT) was introduced to analyze IQAs during the process of establishing regulatory specifications and facilitating continuous manufacturing improvement. Although PAT was introduced in the pharmaceutical industry in the early 21st century, new PAT tools have been introduced during the last 20 years. In this review, we present the recent pharmaceutical PAT tools and their application in pharmaceutical unit operations. Based on unit operations, the significant IQAs monitored by PAT are presented to establish a control strategy for CPV and real time release testing (RTRT). In addition, the equipment type used in unit operation, PAT tools, multivariate statistical tools, and mathematical preprocessing are introduced, along with relevant literature. This review suggests that various PAT tools are rapidly advancing, and various IQAs are efficiently and precisely monitored in the pharmaceutical industry. Therefore, PAT could be a fundamental tool for the present QbD and CPV to improve drug product quality.
Collapse
Affiliation(s)
- Eun Ji Kim
- Department of Pharmaceutical Engineering, Inje University, Gimhae-si, Gyeongnam 621-749, Korea; (E.J.K.); (J.H.K.)
| | - Ji Hyeon Kim
- Department of Pharmaceutical Engineering, Inje University, Gimhae-si, Gyeongnam 621-749, Korea; (E.J.K.); (J.H.K.)
| | - Min-Soo Kim
- College of Pharmacy, Pusan National University, Busandaehak-ro 63 heon-gil, Geumjeong-gu, Busan 46241, Korea;
| | - Seong Hoon Jeong
- College of Pharmacy, Dongguk University-Seoul, Dongguk-ro-32, Ilsan-Donggu, Goyang 10326, Korea;
| | - Du Hyung Choi
- Department of Pharmaceutical Engineering, Inje University, Gimhae-si, Gyeongnam 621-749, Korea; (E.J.K.); (J.H.K.)
| |
Collapse
|