1
|
Huanbutta K, Burapapadh K, Kraisit P, Sriamornsak P, Ganokratanaa T, Suwanpitak K, Sangnim T. Artificial intelligence-driven pharmaceutical industry: A paradigm shift in drug discovery, formulation development, manufacturing, quality control, and post-market surveillance. Eur J Pharm Sci 2024; 203:106938. [PMID: 39419129 DOI: 10.1016/j.ejps.2024.106938] [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: 06/28/2024] [Revised: 10/01/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
The advent of artificial intelligence (AI) has catalyzed a profound transformation in the pharmaceutical industry, ushering in a paradigm shift across various domains, including drug discovery, formulation development, manufacturing, quality control, and post-market surveillance. This comprehensive review examines the multifaceted impact of AI-driven technologies on all stages of the pharmaceutical life cycle. It discusses the application of machine learning algorithms, data analytics, and predictive modeling to accelerate drug discovery processes, optimize formulation development, enhance manufacturing efficiency, ensure stringent quality control measures, and revolutionize post-market surveillance methodologies. By describing the advancements, challenges, and future prospects of harnessing AI in the pharmaceutical landscape, this review offers valuable insights into the evolving dynamics of drug development and regulatory practices in the era of AI-driven innovation.
Collapse
Affiliation(s)
- Kampanart Huanbutta
- Department of Manufacturing Pharmacy, College of Pharmacy, Rangsit University, Pathum Thani 12000, Thailand
| | - Kanokporn Burapapadh
- Department of Manufacturing Pharmacy, College of Pharmacy, Rangsit University, Pathum Thani 12000, Thailand
| | - Pakorn Kraisit
- Thammasat University Research Unit in Smart Materials and Innovative Technology for Pharmaceutical Applications (SMIT-Pharm), Faculty of Pharmacy, Thammasat University, Pathumthani 12120, Thailand
| | - Pornsak Sriamornsak
- Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand; Academy of Science, The Royal Society of Thailand, Bangkok, 10300, Thailand
| | - Thittaporn Ganokratanaa
- Applied Computer Science Program, King Mongkut's University of Technology Thonburi, Bangkok, 10140, Thailand
| | - Kittipat Suwanpitak
- Faculty of Pharmaceutical Sciences, Burapha University, 169, Seansook, Muang, Chonburi, 20131, Thailand
| | - Tanikan Sangnim
- Faculty of Pharmaceutical Sciences, Burapha University, 169, Seansook, Muang, Chonburi, 20131, Thailand.
| |
Collapse
|
2
|
Diószegi A, Ficzere M, Mészáros LA, Péterfi O, Farkas A, Galata DL, Nagy ZK. Automated tablet defect detection and the prediction of disintegration time and crushing strength with deep learning based on tablet surface images. Int J Pharm 2024; 667:124896. [PMID: 39489389 DOI: 10.1016/j.ijpharm.2024.124896] [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: 08/05/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
This paper presents novel measurement methods, where deep learning was used to detect tableting defects and determine the crushing strength and disintegration time of tablets on images captured by machine vision. Five different classes of defects were used and the accuracy of the real-time defect recognition performed with the deep learning algorithm YOLOv5 was 99.2 %. The system can already match the production capability of tablet presses, with still further room left for improvement. The YOLOv5 algorithm was also used to determine the disintegration time and crushing strength of tablets produced at different compression force settings based on their surface texture. With these accurate, low-cost methods, the 100 % screening of the produced tablets could be carried out, resulting in the improvement of quality control and effectiveness of pharmaceutical production.
Collapse
Affiliation(s)
- Anna Diószegi
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rakpart 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 rakpart 3, H-1111 Budapest, Hungary
| | - Lilla Alexandra Mészáros
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rakpart 3, H-1111 Budapest, Hungary
| | - Orsolya Péterfi
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rakpart 3, H-1111 Budapest, Hungary
| | - Attila Farkas
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rakpart 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 rakpart 3, H-1111 Budapest, Hungary.
| | - Zsombor Kristóf Nagy
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rakpart 3, H-1111 Budapest, Hungary
| |
Collapse
|
3
|
Gürsoy E, Kaya Y. Brain-GCN-Net: Graph-Convolutional Neural Network for brain tumor identification. Comput Biol Med 2024; 180:108971. [PMID: 39106672 DOI: 10.1016/j.compbiomed.2024.108971] [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: 10/24/2023] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 08/09/2024]
Abstract
BACKGROUND The intersection of artificial intelligence and medical image analysis has ushered in a new era of innovation and changed the landscape of brain tumor detection and diagnosis. Correct detection and classification of brain tumors based on medical images is crucial for early diagnosis and effective treatment. Convolutional Neural Network (CNN) models are widely used for disease detection. However, they are sometimes unable to sufficiently recognize the complex features of medical images. METHODS This paper proposes a fused Deep Learning (DL) model that combines Graph Neural Networks (GNN), which recognize relational dependencies of image regions, and CNN, which captures spatial features, is proposed to improve brain tumor detection. By integrating these two architectures, our model achieves a more comprehensive representation of brain tumor images and improves classification performance. The proposed model is evaluated on a public dataset of 10847 MRI images. The results show that the proposed model outperforms the existing pre-trained models and traditional CNN architectures. RESULTS The fused DL model achieves 93.68% accuracy in brain tumor classification. The results indicate that the proposed model outperforms the existing pre-trained models and traditional CNN architectures. CONCLUSION The numerical results suggest that the model should be further investigated for potential use in clinical trials to improve clinical decision-making.
Collapse
Affiliation(s)
- Ercan Gürsoy
- Department of Computer Engineering, Adana Alparslan Turkes Science and Technology University, Adana, 01250, Turkey.
| | - Yasin Kaya
- Department of Artificial Intelligence Engineering, Adana Alparslan Turkes Science and Technology University, Adana, 01250, Turkey.
| |
Collapse
|
4
|
Lin HC, Xiao SX. Achievement of Dynamic Tablet Defect Detection Mechanism Using Biaxial Slope Symmetry Algorithm. J Pharm Sci 2024; 113:2208-2214. [PMID: 38484876 DOI: 10.1016/j.xphs.2024.03.004] [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: 12/24/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/24/2024]
Abstract
An inspection in tablet appearance integrity before bottling is regarded as a routine task in a pharmaceutical factory. Although some methods such as automated optical instrument, video or artificial intelligence (AI) are currently available in industry, it usually pays for a complex computational process as well as high cost. Based on the symmetry of tablet appearance in reality, this study develops a biaxial scanning slope symmetry algorithm to realize a dynamic real-time tablet defect detection with a simple arithmetic operation. First, the tablet is discretely scanned using image sensor in two axes, i.e. X and Y directions, simultaneously. Second, the analogy output signals generated from the sensor during the scanning process is discretely digitized and stored in an array. Third, the coordinate of center point in the series data array is identified from every line scanning. Fourth, every section slope between two nearby center points from the first to last lines is formulated and calculated sequentially. Finally, the square mean error (SME) is used to evaluate the shape defect situation according to all accumulated errors from every slope variation. The experimental results verify that the proposed algorithm can achieve both fast and accurate detection performance.
Collapse
Affiliation(s)
- Hsiung-Cheng Lin
- Department of Electronic Engineering, National Chin-Yi University of Technology, Taichung, Taiwan.
| | - Sheng-Xi Xiao
- Department of Electronic Engineering, National Chin-Yi University of Technology, Taichung, Taiwan
| |
Collapse
|
5
|
Gholap AD, Uddin MJ, Faiyazuddin M, Omri A, Gowri S, Khalid M. Advances in artificial intelligence for drug delivery and development: A comprehensive review. Comput Biol Med 2024; 178:108702. [PMID: 38878397 DOI: 10.1016/j.compbiomed.2024.108702] [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: 01/03/2024] [Revised: 05/12/2024] [Accepted: 06/01/2024] [Indexed: 07/24/2024]
Abstract
Artificial intelligence (AI) has emerged as a powerful tool to revolutionize the healthcare sector, including drug delivery and development. This review explores the current and future applications of AI in the pharmaceutical industry, focusing on drug delivery and development. It covers various aspects such as smart drug delivery networks, sensors, drug repurposing, statistical modeling, and simulation of biotechnological and biological systems. The integration of AI with nanotechnologies and nanomedicines is also examined. AI offers significant advancements in drug discovery by efficiently identifying compounds, validating drug targets, streamlining drug structures, and prioritizing response templates. Techniques like data mining, multitask learning, and high-throughput screening contribute to better drug discovery and development innovations. The review discusses AI applications in drug formulation and delivery, clinical trials, drug safety, and pharmacovigilance. It addresses regulatory considerations and challenges associated with AI in pharmaceuticals, including privacy, data security, and interpretability of AI models. The review concludes with future perspectives, highlighting emerging trends, addressing limitations and biases in AI models, and emphasizing the importance of collaboration and knowledge sharing. It provides a comprehensive overview of AI's potential to transform the pharmaceutical industry and improve patient care while identifying further research and development areas.
Collapse
Affiliation(s)
- Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar, Maharashtra, 401404, India.
| | - Md Jasim Uddin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Md Faiyazuddin
- School of Pharmacy, Al-Karim University, Katihar, Bihar, 854106, India; Centre for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Tamil Nadu, India.
| | - Abdelwahab Omri
- Department of Chemistry and Biochemistry, The Novel Drug and Vaccine Delivery Systems Facility, Laurentian University, Sudbury, ON, P3E 2C6, Canada.
| | - S Gowri
- PG & Research, Department of Physics, Cauvery College for Women, Tiruchirapalli, Tamil Nadu, 620018, India
| | - Mohammad Khalid
- James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK; Sunway Centre for Electrochemical Energy and Sustainable Technology (SCEEST), School of Engineering and Technology, Sunway University, No. 5, Jalan Universiti, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia; University Centre for Research and Development, Chandigarh University, Mohali, Punjab, 140413, India.
| |
Collapse
|
6
|
Iwata H, Hayashi Y, Koyama T, Hasegawa A, Ohgi K, Kobayashi I, Okuno Y. Feature extraction of particle morphologies of pharmaceutical excipients from scanning electron microscope images using convolutional neural networks. Int J Pharm 2024; 653:123873. [PMID: 38336179 DOI: 10.1016/j.ijpharm.2024.123873] [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: 10/23/2023] [Revised: 01/08/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
Scanning electron microscopy (SEM) images are the most widely used tool for evaluating particle morphology; however, quantitative evaluation using SEM images is time-consuming and often neglected. In this study, we aimed to extract features related to particle morphology of pharmaceutical excipients from SEM images using a convolutional neural network (CNN). SEM images of 67 excipients were acquired and used as models. A classification CNN model of the excipients was constructed based on the SEM images. Further, features were extracted from the middle layer of this CNN model, and the data was compressed to two dimensions using uniform manifold approximation and projection. Lastly, hierarchical clustering analysis (HCA) was performed to categorize the excipients into several clusters and identify similarities among the samples. The classification CNN model showed high accuracy, allowing each excipient to be identified with a high degree of accuracy. HCA revealed that the 67 excipients were classified into seven clusters. Additionally, the particle morphologies of excipients belonging to the same cluster were found to be very similar. These results suggest that CNN models are useful tools for extracting information and identifying similarities among the particle morphologies of excipients.
Collapse
Affiliation(s)
- Hiroaki Iwata
- Graduate School of Medicine, Kyoto University, 53 Shogoin-kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Yoshihiro Hayashi
- Graduate School of Medicine, Kyoto University, 53 Shogoin-kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan; Pharmaceutical Technology Management Department, Production Division, Nichi-Iko Pharmaceutical Co., Ltd., 205-1, Shimoumezawa Namerikawa-shi, Toyama 936-0857, Japan.
| | - Takuto Koyama
- Graduate School of Medicine, Kyoto University, 53 Shogoin-kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Aki Hasegawa
- Graduate School of Medicine, Kyoto University, 53 Shogoin-kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kosuke Ohgi
- Formulation Development Department, Development & Planning Division, Nichi-Iko Pharmaceutical Co., Ltd., 205-1, Shimoumezawa Namerikawa-shi, Toyama 936-0857, Japan
| | - Ippei Kobayashi
- Formulation Development Department, Development & Planning Division, Nichi-Iko Pharmaceutical Co., Ltd., 205-1, Shimoumezawa Namerikawa-shi, Toyama 936-0857, Japan
| | - Yasushi Okuno
- Graduate School of Medicine, Kyoto University, 53 Shogoin-kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan; RIKEN Center for Computational Science, Kobe 650-0047, Japan
| |
Collapse
|
7
|
Waldner S, Wendelspiess E, Detampel P, Schlepütz CM, Huwyler J, Puchkov M. Advanced analysis of disintegrating pharmaceutical compacts using deep learning-based segmentation of time-resolved micro-tomography images. Heliyon 2024; 10:e26025. [PMID: 38384517 PMCID: PMC10878950 DOI: 10.1016/j.heliyon.2024.e26025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 02/23/2024] Open
Abstract
The mechanism governing pharmaceutical tablet disintegration is far from fully understood. Despite the importance of controlling a formulation's disintegration process to maximize the active pharmaceutical ingredient's bioavailability and ensure predictable and consistent release profiles, the current understanding of the process is based on indirect or superficial measurements. Formulation science could, therefore, additionally deepen the understanding of the fundamental physical principles governing disintegration based on direct observations of the process. We aim to help bridge the gap by generating a series of time-resolved X-ray micro-computed tomography (μCT) images capturing volumetric images of a broad range of mini-tablet formulations undergoing disintegration. Automated image segmentation was a prerequisite to overcoming the challenges of analyzing multiple time series of heterogeneous tomographic images at high magnification. We devised and trained a convolutional neural network (CNN) based on the U-Net architecture for autonomous, rapid, and consistent image segmentation. We created our own μCT data reconstruction pipeline and parameterized it to deliver image quality optimal for our CNN-based segmentation. Our approach enabled us to visualize the internal microstructures of the tablets during disintegration and to extract parameters of disintegration kinetics from the time-resolved data. We determine by factor analysis the influence of the different formulation components on the disintegration process in terms of both qualitative and quantitative experimental responses. We relate our findings to known formulation component properties and established experimental results. Our direct imaging approach, enabled by deep learning-based image processing, delivers new insights into the disintegration mechanism of pharmaceutical tablets.
Collapse
Affiliation(s)
- Samuel Waldner
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, Klingelberstrasse 50, 4056, Basel, Switzerland
| | - Erwin Wendelspiess
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, Klingelberstrasse 50, 4056, Basel, Switzerland
| | - Pascal Detampel
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, Klingelberstrasse 50, 4056, Basel, Switzerland
| | | | - Jörg Huwyler
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, Klingelberstrasse 50, 4056, Basel, Switzerland
| | - Maxim Puchkov
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Technology, University of Basel, Klingelberstrasse 50, 4056, Basel, Switzerland
| |
Collapse
|
8
|
Bak A, Burlage R, Greene N, Nambiar P, Lu X, Templeton A. Accelerating Drug Product Development and Approval: Early Development and Evaluation. Pharm Res 2024; 41:1-6. [PMID: 37552384 DOI: 10.1007/s11095-023-03566-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/12/2023] [Indexed: 08/09/2023]
Affiliation(s)
- Annette Bak
- Advanced Drug Delivery, Pharmaceutical Sciences, Biopharmaceutical R&D, AstraZeneca, Boston, MA, USA.
| | - Rubi Burlage
- Pharmaceutical Sciences and Clinical Supplies, Merck & Co, Rahway, NJ, USA
| | - Nigel Greene
- Data Science, Clinical Pharmacology and Safety Sciences, Biopharmaceutical R&D, AstraZeneca, Boston, MA, USA
| | - Prabu Nambiar
- Principal, Syner-G Biopharma Group, Framingham, MA, USA
| | - Xiuling Lu
- Department of Pharmaceutical Sciences, University of Connecticut, School of Pharmacy, Storrs, CT, USA
| | - Allen Templeton
- Pharmaceutical Sciences and Clinical Supplies, Merck & Co, Rahway, NJ, USA
| |
Collapse
|
9
|
Sahu A, Rathee S, Saraf S, Jain SK. A Review on the Recent Advancements and Artificial Intelligence in Tablet Technology. Curr Drug Targets 2024; 25:416-430. [PMID: 38213164 DOI: 10.2174/0113894501281290231221053939] [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: 10/10/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND Tablet formulation could be revolutionized by the integration of modern technology and established pharmaceutical sciences. The pharmaceutical sector can develop tablet formulations that are not only more efficient and stable but also patient-friendly by utilizing artificial intelligence (AI), machine learning (ML), and materials science. OBJECTIVES The primary objective of this review is to explore the advancements in tablet technology, focusing on the integration of modern technologies like artificial intelligence (AI), machine learning (ML), and materials science to enhance the efficiency, cost-effectiveness, and quality of tablet formulation processes. METHODS This review delves into the utilization of AI and ML techniques within pharmaceutical research and development. The review also discusses various ML methodologies employed, including artificial neural networks, an ensemble of regression trees, support vector machines, and multivariate data analysis techniques. RESULTS Recent studies showcased in this review demonstrate the feasibility and effectiveness of ML approaches in pharmaceutical research. The application of AI and ML in pharmaceutical research has shown promising results, offering a potential avenue for significant improvements in the product development process. CONCLUSION The integration of nanotechnology, AI, ML, and materials science with traditional pharmaceutical sciences presents a remarkable opportunity for enhancing tablet formulation processes. This review collectively underscores the transformative role that AI and ML can play in advancing pharmaceutical research and development, ultimately leading to more efficient, reliable and patient-centric tablet formulations.
Collapse
Affiliation(s)
- Amit Sahu
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Sunny Rathee
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Shivani Saraf
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Sanjay K Jain
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| |
Collapse
|
10
|
Jiang J, Lu A, Ma X, Ouyang D, Williams RO. The applications of machine learning to predict the forming of chemically stable amorphous solid dispersions prepared by hot-melt extrusion. Int J Pharm X 2023; 5:100164. [PMID: 36798832 PMCID: PMC9925947 DOI: 10.1016/j.ijpx.2023.100164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
Amorphous solid dispersion (ASD) is one of the most important strategies to improve the solubility and dissolution rate of poorly water-soluble drugs. As a widely used technique to prepare ASDs, hot-melt extrusion (HME) provides various benefits, including a solvent-free process, continuous manufacturing, and efficient mixing compared to solvent-based methods, such as spray drying. Energy input, consisting of thermal and specific mechanical energy, should be carefully controlled during the HME process to prevent chemical degradation and residual crystallinity. However, a conventional ASD development process uses a trial-and-error approach, which is laborious and time-consuming. In this study, we have successfully built multiple machine learning (ML) models to predict the amorphization of crystalline drug formulations and the chemical stability of subsequent ASDs prepared by the HME process. We utilized 760 formulations containing 49 active pharmaceutical ingredients (APIs) and multiple types of excipients. By evaluating the built ML models, we found that ECFP-LightGBM was the best model to predict amorphization with an accuracy of 92.8%. Furthermore, ECFP-XGBoost was the best in estimating chemical stability with an accuracy of 96.0%. In addition, the feature importance analyses based on SHapley Additive exPlanations (SHAP) and information gain (IG) revealed that several processing parameters and material attributes (i.e., drug loading, polymer ratio, drug's Extended-connectivity fingerprints (ECFP) fingerprints, and polymer's properties) are critical for achieving accurate predictions for the selected models. Moreover, important API's substructures related to amorphization and chemical stability were determined, and the results are largely consistent with the literature. In conclusion, we established the ML models to predict formation of chemically stable ASDs and identify the critical attributes during HME processing. Importantly, the developed ML methodology has the potential to facilitate the product development of ASDs manufactured by HME with a much reduced human workload.
Collapse
Affiliation(s)
- Junhuang Jiang
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Anqi Lu
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Xiangyu Ma
- Global Investment Research, Goldman Sachs, NY 10282, USA
| | - Defang Ouyang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, 999078, Macau
| | - Robert O. Williams
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
11
|
Ficzere M, Péterfi O, Farkas A, Nagy ZK, Galata DL. Image-based simultaneous particle size distribution and concentration measurement of powder blend components with deep learning and machine vision. Eur J Pharm Sci 2023; 191:106611. [PMID: 37844806 DOI: 10.1016/j.ejps.2023.106611] [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: 04/11/2023] [Revised: 08/21/2023] [Accepted: 10/14/2023] [Indexed: 10/18/2023]
Abstract
This work presents a system, where deep learning was used on images captured with a digital camera to simultaneously determine the API concentration and the particle size distribution (PSD) of two components of a powder blend. The blend consisted of acetylsalicylic acid (ASA) and calcium hydrogen phosphate (CHP), and the predicted API concentration was found corresponding with the HPLC measurements. The PSDs determined with the method corresponded with those measured with laser diffraction particle size analysis. This novel method provides fast and simple measurements and could be suitable for detecting segregation in the powder. By examining the powders discharged from a batch blender, the API concentrations at the top and bottom of the container could be measured, yielding information about the adequacy of the blending and improving the quality control of the manufacturing process.
Collapse
Affiliation(s)
- Máté Ficzere
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp 3., Budapest H 1111, Hungary
| | - Orsolya Péterfi
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp 3., Budapest H 1111, Hungary
| | - Attila Farkas
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp 3., Budapest H 1111, Hungary
| | - Zsombor Kristóf Nagy
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp 3., Budapest H 1111, 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., Budapest H 1111, Hungary
| |
Collapse
|
12
|
Xu H, Wu L, Xue Y, Yang T, Xiong T, Wang C, He S, Sun H, Cao Z, Liu J, Wang S, Li Z, Naeem A, Yin X, Zhang J. Advances in Structure Pharmaceutics from Discovery to Evaluation and Design. Mol Pharm 2023; 20:4404-4429. [PMID: 37552597 DOI: 10.1021/acs.molpharmaceut.3c00514] [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: 08/10/2023]
Abstract
Drug delivery systems (DDSs) play an important role in delivering active pharmaceutical ingredients (APIs) to targeted sites with a predesigned release pattern. The chemical and biological properties of APIs and excipients have been extensively studied for their contribution to DDS quality and effectiveness; however, the structural characteristics of DDSs have not been adequately explored. Structure pharmaceutics involves the study of the structure of DDSs, especially the three-dimensional (3D) structures, and its interaction with the physiological and pathological structure of organisms, possibly influencing their release kinetics and targeting abilities. A systematic overview of the structures of a variety of dosage forms, such as tablets, granules, pellets, microspheres, powders, and nanoparticles, is presented. Moreover, the influence of structures on the release and targeting capability of DDSs has also been discussed, especially the in vitro and in vivo release correlation and the structure-based organ- and tumor-targeting capabilities of particles with different structures. Additionally, an in-depth discussion is provided regarding the application of structural strategies in the DDSs design and evaluation. Furthermore, some of the most frequently used characterization techniques in structure pharmaceutics are briefly described along with their potential future applications.
Collapse
Affiliation(s)
- Huipeng Xu
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Wu
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, School of Pharmacy, Ministry of Education, Yantai University, Yantai 264005, China
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Yanling Xue
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China
| | - Ting Yang
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ting Xiong
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Caifen Wang
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Siyu He
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongyu Sun
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zeying Cao
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun Liu
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Siwen Wang
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhe Li
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Abid Naeem
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Xianzhen Yin
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Lingang Laboratory, Shanghai 201602, China
| | - Jiwen Zhang
- Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, National Institutes for Food and Drug Control, No.2 Tiantan Xili, Beijing 100050, China
| |
Collapse
|
13
|
Taseva AR, Persoons T, D'Arcy DM. Application of an AI image analysis and classification approach to characterise dissolution and precipitation events in the flow through apparatus. Eur J Pharm Biopharm 2023; 189:36-47. [PMID: 37120067 DOI: 10.1016/j.ejpb.2023.04.020] [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: 12/20/2022] [Revised: 04/19/2023] [Accepted: 04/22/2023] [Indexed: 05/01/2023]
Abstract
Imaging and artificial intelligence (AI) approaches have been used with increasing frequency in pharmaceutical industry in recent years. Characterisation of processes such as drug dissolution and precipitation is vital in quality control testing and drug manufacture. To support existing techniques like in vitro dissolution testing, novel process analytical technologies (PATs) can give an insight into these processes. The aim of this study was to create and explore the potential of an automated image classification model based on image analysis to identify events (dissolution and precipitation) occurring in the flow-through apparatus (FTA) test cell, and the ability to characterise a dissolution process over time. Several precipitation conditions were tested in a USP 4 FTA test cell with images recorded during early (plume formation) and late (particulate re-formation) stages of precipitation. An available MATLAB code was used as a base to develop and validate an anomaly classification model able to detect different events occurring during the precipitation process in the dissolution cell. Two variants of the model were tested on images from a dissolution test in the FTA, with a view to application of the image analysis system to quantitative characterization of the dissolution process over time. It was found that the classification model is highly accurate (>90%) in detecting events occurring in the FTA test cell. The model showed potential to be used to characterise the stages of dissolution and precipitation processes, and as a proof of concept demonstrates potential for deep machine learning image analysis to be applied to kinetics of other pharmaceutical processes.
Collapse
Affiliation(s)
- Alexandra R Taseva
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland; SSPC, The Science Foundation Ireland Research Centre for Pharmaceuticals, Trinity College Dublin, Ireland.
| | - Tim Persoons
- Department of Mechanical, Manufacturing & Biomedical Engineering, Trinity College Dublin, Ireland; SSPC, The Science Foundation Ireland Research Centre for Pharmaceuticals, Trinity College Dublin, Ireland.
| | - Deirdre M D'Arcy
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland; SSPC, The Science Foundation Ireland Research Centre for Pharmaceuticals, Trinity College Dublin, Ireland.
| |
Collapse
|
14
|
Vora LK, Gholap AD, Jetha K, Thakur RRS, Solanki HK, Chavda VP. Artificial Intelligence in Pharmaceutical Technology and Drug Delivery Design. Pharmaceutics 2023; 15:1916. [PMID: 37514102 PMCID: PMC10385763 DOI: 10.3390/pharmaceutics15071916] [Citation(s) in RCA: 61] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/28/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Artificial intelligence (AI) has emerged as a powerful tool that harnesses anthropomorphic knowledge and provides expedited solutions to complex challenges. Remarkable advancements in AI technology and machine learning present a transformative opportunity in the drug discovery, formulation, and testing of pharmaceutical dosage forms. By utilizing AI algorithms that analyze extensive biological data, including genomics and proteomics, researchers can identify disease-associated targets and predict their interactions with potential drug candidates. This enables a more efficient and targeted approach to drug discovery, thereby increasing the likelihood of successful drug approvals. Furthermore, AI can contribute to reducing development costs by optimizing research and development processes. Machine learning algorithms assist in experimental design and can predict the pharmacokinetics and toxicity of drug candidates. This capability enables the prioritization and optimization of lead compounds, reducing the need for extensive and costly animal testing. Personalized medicine approaches can be facilitated through AI algorithms that analyze real-world patient data, leading to more effective treatment outcomes and improved patient adherence. This comprehensive review explores the wide-ranging applications of AI in drug discovery, drug delivery dosage form designs, process optimization, testing, and pharmacokinetics/pharmacodynamics (PK/PD) studies. This review provides an overview of various AI-based approaches utilized in pharmaceutical technology, highlighting their benefits and drawbacks. Nevertheless, the continued investment in and exploration of AI in the pharmaceutical industry offer exciting prospects for enhancing drug development processes and patient care.
Collapse
Affiliation(s)
- Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Keshava Jetha
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
- Ph.D. Section, Gujarat Technological University, Ahmedabad 382424, Gujarat, India
| | | | - Hetvi K Solanki
- Pharmacy Section, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| |
Collapse
|
15
|
Lee D, Weinhardt F, Hommel J, Piotrowski J, Class H, Steeb H. Machine learning assists in increasing the time resolution of X-ray computed tomography applied to mineral precipitation in porous media. Sci Rep 2023; 13:10529. [PMID: 37386125 DOI: 10.1038/s41598-023-37523-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/22/2023] [Indexed: 07/01/2023] Open
Abstract
Many subsurface engineering technologies or natural processes cause porous medium properties, such as porosity or permeability, to evolve in time. Studying and understanding such processes on the pore scale is strongly aided by visualizing the details of geometric and morphological changes in the pores. For realistic 3D porous media, X-Ray Computed Tomography (XRCT) is the method of choice for visualization. However, the necessary high spatial resolution requires either access to limited high-energy synchrotron facilities or data acquisition times which are considerably longer (e.g. hours) than the time scales of the processes causing the pore geometry change (e.g. minutes). Thus, so far, conventional benchtop XRCT technologies are often too slow to allow for studying dynamic processes. Interrupting experiments for performing XRCT scans is also in many instances no viable approach. We propose a novel workflow for investigating dynamic precipitation processes in porous media systems in 3D using a conventional XRCT technology. Our workflow is based on limiting the data acquisition time by reducing the number of projections and enhancing the lower-quality reconstructed images using machine-learning algorithms trained on images reconstructed from high-quality initial- and final-stage scans. We apply the proposed workflow to induced carbonate precipitation within a porous-media sample of sintered glass-beads. So we were able to increase the temporal resolution sufficiently to study the temporal evolution of the precipitate accumulation using an available benchtop XRCT device.
Collapse
Affiliation(s)
- Dongwon Lee
- Institute of Applied Mechanics (CE), University of Stuttgart, Pfaffenwaldring 7, 70569, Stuttgart, Germany.
| | - Felix Weinhardt
- Institute for Modelling Hydraulic and Environmental Systems, University of Stuttgart, Pfaffenwaldring 61, 70569, Stuttgart, Germany
| | - Johannes Hommel
- Institute for Modelling Hydraulic and Environmental Systems, University of Stuttgart, Pfaffenwaldring 61, 70569, Stuttgart, Germany
| | - Joseph Piotrowski
- Agrosphere (IBG-3), Institute of Bio- and Geosciences, Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Holger Class
- Institute for Modelling Hydraulic and Environmental Systems, University of Stuttgart, Pfaffenwaldring 61, 70569, Stuttgart, Germany
| | - Holger Steeb
- Institute of Applied Mechanics (CE), University of Stuttgart, Pfaffenwaldring 7, 70569, Stuttgart, Germany
- SC SimTech, University of Stuttgart, Pfaffenwaldring 5, 70569, Stuttgart, Germany
| |
Collapse
|
16
|
Vijayakumar J, Goudarzi NM, Eeckhaut G, Schrijnemakers K, Cnudde V, Boone MN. Characterization of Pharmaceutical Tablets by X-ray Tomography. Pharmaceuticals (Basel) 2023; 16:ph16050733. [PMID: 37242516 DOI: 10.3390/ph16050733] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/07/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Solid dosage forms such as tablets are extensively used in drug administration for their simplicity and large-scale manufacturing capabilities. High-resolution X-ray tomography is one of the most valuable non-destructive techniques to investigate the internal structure of the tablets for drug product development as well as for a cost effective production process. In this work, we review the recent developments in high-resolution X-ray microtomography and its application towards different tablet characterizations. The increased availability of powerful laboratory instrumentation, as well as the advent of high brilliance and coherent 3rd generation synchrotron light sources, combined with advanced data processing techniques, are driving the application of X-ray microtomography forward as an indispensable tool in the pharmaceutical industry.
Collapse
Affiliation(s)
- Jaianth Vijayakumar
- Centre for X-ray Tomography (UGCT), Ghent University, Proeftuinstraat 86/N3, 9000 Gent, Belgium
- Department of Physics and Astronomy, Radiation Physics, Ghent University, Proeftuinstraat 86/N12, 9000 Gent, Belgium
| | - Niloofar Moazami Goudarzi
- Centre for X-ray Tomography (UGCT), Ghent University, Proeftuinstraat 86/N3, 9000 Gent, Belgium
- Department of Physics and Astronomy, Radiation Physics, Ghent University, Proeftuinstraat 86/N12, 9000 Gent, Belgium
| | - Guy Eeckhaut
- Janssen Pharmaceutica, Turnhoutseweg 30, 2340 Beerse, Belgium
| | | | - Veerle Cnudde
- Centre for X-ray Tomography (UGCT), Ghent University, Proeftuinstraat 86/N3, 9000 Gent, Belgium
- Pore-Scale Processes in Geomaterials Research (PProGRess), Department of Geology, Ghent University, Krijgslaan 281/S8, 9000 Gent, Belgium
- Environmental Hydrogeology, Department of Earth Sciences, Faculty of Geosciences, Utrecht University, Princetonlaan 8A, 3584 CD Utrecht, The Netherlands
| | - Matthieu N Boone
- Centre for X-ray Tomography (UGCT), Ghent University, Proeftuinstraat 86/N3, 9000 Gent, Belgium
- Department of Physics and Astronomy, Radiation Physics, Ghent University, Proeftuinstraat 86/N12, 9000 Gent, Belgium
| |
Collapse
|
17
|
Jiang J, Ouyang D, Williams RO. Predicting Glass-Forming Ability of Pharmaceutical Compounds by Using Machine Learning Technologies. AAPS PharmSciTech 2023; 24:103. [PMID: 37072563 DOI: 10.1208/s12249-023-02535-6] [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: 11/30/2022] [Accepted: 02/14/2023] [Indexed: 04/20/2023] Open
Abstract
Low aqueous solubility is a common and serious challenge for most drug substances not only in development but also in the market, and it may cause low absorption and bioavailability as a result. Amorphization is an intermolecular modification strategy to address the issue by breaking the crystal lattice and enhancing the energy state. However, due to the physicochemical properties of the amorphous state, drugs are thermodynamically unstable and tend to recrystallize over time. Glass-forming ability (GFA) is an experimental method to evaluate the forming and stability of glass formed by crystallization tendency. Machine learning (ML) is an emerging technique widely applied in pharmaceutical sciences. In this study, we successfully developed multiple ML models (i.e., random forest (RF), XGBoost, and support vector machine (SVM)) to predict GFA from 171 drug molecules. Two different molecular representation methods (i.e., 2D descriptor and Extended-connectivity fingerprints (ECFP)) were implemented to process the drug molecules. Among all ML algorithms, 2D-RF performed best with the highest accuracy, AUC, and F1 of 0.857, 0.850, and 0.828, respectively, in the testing set. In addition, we conducted a feature importance analysis, and the results mostly agreed with the literature, which demonstrated the interpretability of the model. Most importantly, our study showed great potential for developing amorphous drugs by in silico screening of stable glass formers.
Collapse
Affiliation(s)
- Junhuang Jiang
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, Texas, 78712, USA
| | - Defang Ouyang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, Macau, 999078, China
| | - Robert O Williams
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, Texas, 78712, USA.
| |
Collapse
|
18
|
Sultan T, Dave VS, Cetinkaya C. Early detection and assessment of invisible cracks in compressed oral solid dosage forms. Int J Pharm 2023; 635:122786. [PMID: 36854370 DOI: 10.1016/j.ijpharm.2023.122786] [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: 11/27/2022] [Revised: 02/17/2023] [Accepted: 02/24/2023] [Indexed: 03/01/2023]
Abstract
In the pharmaceutical manufacturing industry, real-time in situ quality monitoring for detecting defects at an early stage is a desirable ability, especially in high-rate production, to minimize downstream quality-related issues, financial losses, and timeline risks. In this study, we focus on the early detection of crack formation in compressed oral solid dosage (OSD) forms at its onset before complete delamination and/or capping in downstream processing. The detection of internal tablet cracks related to local micro-stress/strain states, internal granularity (texture), and micro-structure failures is rather unlikely by traditional testing methods, such as the USP reference standards for friability, fracturing, or hardness testing. In addition, these tests do not permit the objective and quantitative evaluation of the influence of formulation and process parameters, which are critical for the development of high-quality drug products manufactured at high rates on a large scale. Internal cracks (potentially resulting in 'capping' and/or 'lamination') under high-strain compaction of highly visco-elastic powder materials are a common failure mode. In the current study, two approaches are introduced and utilized for non-destructively detecting and evaluating hidden cracks in pharmaceutical compacts based on (i) varying axial load-displacement measurements and (ii) ultrasonic reflection ray tracing. The reflection ray tracing technique is a non-destructive, inexpensive, rapid, and material-sparing approach, which makes it advantageous for real-time quality monitoring and defect characterization applications. The varying axial load-displacement technique is more suitable for analytical studies, especially in the design and development phases of compressed OSD products. In this study, as a model application, utilizing these two approaches, it is demonstrated how internal and external cracks can be detected, localized, characterized, and analyzed as a function of disintegrant ratio and main compression force. Various uses of these two techniques in practice, such as in Continuous Manufacturing (CM) and Real-Time Release Testing (RTRT), are also discussed.
Collapse
Affiliation(s)
- Tipu Sultan
- Photo-Acoustics Research Laboratory Department of Mechanical and Aerospace Engineering, Clarkson University Potsdam, NY 13699-5725, USA
| | - Vivek S Dave
- St. John Fisher University, Wegmans School of Pharmacy, Rochester, NY 14618, USA
| | - Cetin Cetinkaya
- Photo-Acoustics Research Laboratory Department of Mechanical and Aerospace Engineering, Clarkson University Potsdam, NY 13699-5725, USA.
| |
Collapse
|
19
|
Predicting mini-tablet dissolution performance utilizing X-ray computed tomography. Eur J Pharm Sci 2023; 181:106346. [PMID: 36494000 DOI: 10.1016/j.ejps.2022.106346] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
Mini-tablets (MTs) have been utilized as an alternative to monolithic tablets due to their ease of use for pediatric populations, dose flexibility and tailoring of drug release profiles. Similar to monolithic tablets, MTs can develop film coat and internal core defects during manufacturing processes that may adversely affect their dissolution performance. The use of x-ray computed microtomography (XRCT) is well documented for monolithic tablets as a means of identifying internal defects, but applications to MTs have not been well studied. In this study, we have developed a workflow that analyzes reconstructed XRCT images of enteric-coated mini-tablets using deep learning convolutional neural networks. This algorithm was utilized to extract key physical features of individual MTs, such as micro-crack volume and enteric coat thickness. By performing dissolution studies on individual MTs, correlations were established based on the physical parameters obtained by XRCT and the dissolution performance, enabling prediction of dissolution performance utilizing non-destructive imaging data. This workflow provides insight into the physical variability of MT populations that are generated during manufacturing, enabling optimization of critical tableting and coating parameters to achieve the target dissolution criteria. Through this mechanistic understanding, quality is built into the final drug product through rational development of formulation and process parameters.
Collapse
|
20
|
Jiang J, Ma X, Ouyang D, Williams RO. Emerging Artificial Intelligence (AI) Technologies Used in the Development of Solid Dosage Forms. Pharmaceutics 2022; 14:2257. [PMID: 36365076 PMCID: PMC9694557 DOI: 10.3390/pharmaceutics14112257] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 07/30/2023] Open
Abstract
Artificial Intelligence (AI)-based formulation development is a promising approach for facilitating the drug product development process. AI is a versatile tool that contains multiple algorithms that can be applied in various circumstances. Solid dosage forms, represented by tablets, capsules, powder, granules, etc., are among the most widely used administration methods. During the product development process, multiple factors including critical material attributes (CMAs) and processing parameters can affect product properties, such as dissolution rates, physical and chemical stabilities, particle size distribution, and the aerosol performance of the dry powder. However, the conventional trial-and-error approach for product development is inefficient, laborious, and time-consuming. AI has been recently recognized as an emerging and cutting-edge tool for pharmaceutical formulation development which has gained much attention. This review provides the following insights: (1) a general introduction of AI in the pharmaceutical sciences and principal guidance from the regulatory agencies, (2) approaches to generating a database for solid dosage formulations, (3) insight on data preparation and processing, (4) a brief introduction to and comparisons of AI algorithms, and (5) information on applications and case studies of AI as applied to solid dosage forms. In addition, the powerful technique known as deep learning-based image analytics will be discussed along with its pharmaceutical applications. By applying emerging AI technology, scientists and researchers can better understand and predict the properties of drug formulations to facilitate more efficient drug product development processes.
Collapse
Affiliation(s)
- Junhuang Jiang
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Xiangyu Ma
- Global Investment Research, Goldman Sachs, New York, NY 10282, USA
| | - Defang Ouyang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, Macau 999078, China
| | - Robert O. Williams
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
21
|
Iwata H, Hayashi Y, Hasegawa A, Terayama K, Okuno Y. Classification of scanning electron microscope images of pharmaceutical excipients using deep convolutional neural networks with transfer learning. Int J Pharm X 2022; 4:100135. [PMID: 36325273 PMCID: PMC9619299 DOI: 10.1016/j.ijpx.2022.100135] [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: 08/25/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
Convolutional Neural Networks (CNNs) are image analysis techniques that have been applied to image classification in various fields. In this study, we applied a CNN to classify scanning electron microscopy (SEM) images of pharmaceutical raw material powders to determine if a CNN can evaluate particle morphology. We tested 10 pharmaceutical excipients with widely different particle morphologies. SEM images for each excipient were acquired and divided into training, validation, and test sets. Classification models were constructed by applying transfer learning to pretrained CNN models such as VGG16 and ResNet50. The results of a 5-fold cross-validation showed that the classification accuracy of the CNN model was sufficiently high using either pretrained model and that the type of excipient could be classified with high accuracy. The results suggest that the CNN model can detect differences in particle morphology, such as particle size, shape, and surface condition. By applying Grad-CAM to the constructed CNN model, we succeeded in finding particularly important regions in the particle image of the excipients. CNNs have been found to have the potential to be applied to the identification and characterization of raw material powders for pharmaceutical development.
Collapse
Affiliation(s)
- Hiroaki Iwata
- Graduate School of Medicine, Kyoto University, 53 Shogoin-kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yoshihiro Hayashi
- Graduate School of Medicine, Kyoto University, 53 Shogoin-kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan,Pharmaceutical Technology Division, Nichi-Iko Pharmaceutical Co., Ltd., 205-1, Shimoumezawa Namerikawa-shi, Toyama 936-0857, Japan,Correspondence to: Y. Hayashi, Pharmaceutical Technology Division, Nichi-Iko Pharmaceutical Co., Ltd.; 205-1, Shimoumezawa Namerikawa-shi, Toyama 936-0857, Japan.
| | - Aki Hasegawa
- Graduate School of Medicine, Kyoto University, 53 Shogoin-kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kei Terayama
- Graduate School of Medical Life Science, Yokohama City University, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Yasushi Okuno
- Graduate School of Medicine, Kyoto University, 53 Shogoin-kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan,RIKEN Center for Computational Science, Kobe 650-0047, Japan,Correspondence to: Y. Okuno, Graduate School of Medicine, Kyoto University, 53 Shogoin-kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
22
|
Jiang J, Peng HH, Yang Z, Ma X, Sahakijpijarn S, Moon C, Ouyang D, Williams Iii RO. The applications of Machine learning (ML) in designing dry powder for inhalation by using thin-film-freezing technology. Int J Pharm 2022; 626:122179. [PMID: 36084876 DOI: 10.1016/j.ijpharm.2022.122179] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 12/19/2022]
Abstract
Dry powder inhalers (DPIs) are one of the most widely used devices for treating respiratory diseases. Thin--film--freezing (TFF) is a particle engineering technology that has been demonstrated to prepare dry powder for inhalation with enhanced physicochemical properties. Aerosol performance, which is indicated by fine particle fraction (FPF) and mass median aerodynamic diameter (MMAD), is an important consideration during the product development process. However, the conventional approach for formulation development requires many trial-and-error experiments, which is both laborious and time consuming. As a state-of-the art technique, machine learning has gained more attention in pharmaceutical science and has been widely applied in different settings. In this study, we have successfully built a prediction model for aerosol performance by using both tabular data and scanning electron microscopy (SEM) images. TFF technology was used to prepare 134 dry powder formulations which were collected as a tabular dataset. After testing many machine learning models, we determined that the Random Forest (RF) model was best for FPF prediction with a mean absolute error of ± 7.251%, and artificial neural networks (ANNs) performed the best in estimating MMAD with a mean absolute error of ± 0.393 μm. In addition, a convolutional neural network was employed for SEM image classification and has demonstrated high accuracy (>83.86%) and adaptability in predicting 316 SEM images of three different drug formulations. In conclusion, the machine learning models using both tabular data and image classification were successfully established to evaluate the aerosol performance of dry powder for inhalation. These machine learning models facilitate the product development process of dry powder for inhalation manufactured by TFF technology and have the potential to significantly reduce the product development workload. The machine learning methodology can also be applied to other formulation design and development processes in the future.
Collapse
Affiliation(s)
- Junhuang Jiang
- Department of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, TX, USA
| | - Han-Hsuan Peng
- Department of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, TX, USA
| | - Zhenpei Yang
- Department of Computer Science, The University of Texas at Austin, TX, USA
| | - Xiangyu Ma
- Global Investment Research, Goldman Sachs, NY, USA
| | | | - Chaeho Moon
- Department of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, TX, USA
| | - Defang Ouyang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, Macau, China
| | - Robert O Williams Iii
- Department of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, TX, USA.
| |
Collapse
|
23
|
Shibata H, Terabe M, Shibano Y, Saitoh S, Takasugi T, Hayashi Y, Okabe S, Yamaguchi Y, Yasukawa H, Suetomo H, Miyanabe K, Ohbayashi N, Akimaru M, Saito S, Ito D, Nakano A, Kojima S, Miyahara Y, Sasaki K, Maruno T, Noda M, Kiyoshi M, Harazono A, Torisu T, Uchiyama S, Ishii-Watabe A. A Collaborative Study on the Classification of Silicone Oil Droplets and Protein Particles Using Flow Imaging Method. J Pharm Sci 2022; 111:2745-2757. [PMID: 35839866 DOI: 10.1016/j.xphs.2022.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 10/17/2022]
Abstract
In this study, we conducted a collaborative study on the classification between silicone oil droplets and protein particles detected using the flow imaging (FI) method toward proposing a standardized classifier/model. We compared four approaches, including a classification filter composed of particle characteristic parameters, principal component analysis, decision tree, and convolutional neural network in the performance of the developed classifier/model. Finally, the points to be considered were summarized for measurement using the FI method, and for establishing the classifier/model using machine learning to differentiate silicone oil droplets and protein particles.
Collapse
Affiliation(s)
- Hiroko Shibata
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan.
| | - Masahiro Terabe
- Pharmaceutical Technology Division, Analytical Development Department, Chugai Pharmaceutical Co. Ltd., 5-1 Ukima, 5-chome, Kita-ku, Tokyo 115-8543 Japan
| | - Yuriko Shibano
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Satoshi Saitoh
- Pharmaceutical Technology Division, Analytical Development Department, Chugai Pharmaceutical Co. Ltd., 5-1 Ukima, 5-chome, Kita-ku, Tokyo 115-8543 Japan
| | - Tomohiro Takasugi
- Analytical Research Laboratories, Pharmaceutical Technology, Astellas Pharma. Inc., 5-2-3 Tokodai, Tsukuba, Ibaraki, 300-2698, Japan
| | - Yu Hayashi
- Analytical Research Laboratories, Pharmaceutical Technology, Astellas Pharma. Inc., 5-2-3 Tokodai, Tsukuba, Ibaraki, 300-2698, Japan
| | - Shinji Okabe
- Research Division, CMC Development Research, Formulation Research Unit, Formulation Development, JCR Pharmaceuticals Co., Ltd., 2-2-9 Murotani, Nishi-ku, Kobe, Hyogo 651-2241, Japan
| | - Yuka Yamaguchi
- Research Division, CMC Development Research, Formulation Research Unit, Formulation Development, JCR Pharmaceuticals Co., Ltd., 2-2-9 Murotani, Nishi-ku, Kobe, Hyogo 651-2241, Japan
| | - Hidehito Yasukawa
- Research Division, CMC Development Research, Formulation Research Unit, Formulation Development, JCR Pharmaceuticals Co., Ltd., 2-2-9 Murotani, Nishi-ku, Kobe, Hyogo 651-2241, Japan
| | - Hiroyuki Suetomo
- Bio Process Research and Development Laboratories, Production Division, Kyowa Kirin Co., Ltd., 100-1, Hagiwara-machi, Takasaki, Gunma 370-0013, Japan
| | - Kazuhiro Miyanabe
- CMC Regulatory and Analytical R&D., Ono Pharmaceutical Co., Ltd., 1-1, Sakurai 3-chome, Shimamoto-cho, Mishima-gun, Osaka, 618-8585, Japan
| | - Naomi Ohbayashi
- Pharmaceutical Research Center, Formulation Research Lab., Meiji Seika Pharma Co., Ltd., 788 Kayama, Odawara, Kanagawa, 250-0852, Japan
| | - Michiko Akimaru
- Analytical & Quality Evaluation Research Laboratories, Daiichi Sankyo Co., Ltd., 1-12-1, Shinomiya, Hiratsuka, Kanagawa, 254-0014, Japan
| | - Shuntaro Saito
- Analytical & Quality Evaluation Research Laboratories, Daiichi Sankyo Co., Ltd., 1-12-1, Shinomiya, Hiratsuka, Kanagawa, 254-0014, Japan
| | - Daisuke Ito
- Japan Blood Products Organization, 1007-31 Izumisawa, Chitose, Hokkaido, 066-8610, Japan
| | - Atsushi Nakano
- Japan Blood Products Organization, 1007-31 Izumisawa, Chitose, Hokkaido, 066-8610, Japan
| | - Shota Kojima
- Pharmaceutical Laboratory, Mochida Pharmaceutical Co., Ltd. 342 Gensuke, Fujieda, Shizuoka, 426-8640, Japan
| | - Yuya Miyahara
- CMC Modality Technology Laboratories, Production Technology & Supply Chain Management Division, Mitsubishi Tanabe Pharma Corporation, 7473-2, Onoda, Sanyoonoda-shi, Yamaguchi, 756-0054 Japan
| | - Kenji Sasaki
- CMC Modality Technology Laboratories, Production Technology & Supply Chain Management Division, Mitsubishi Tanabe Pharma Corporation, 7473-2, Onoda, Sanyoonoda-shi, Yamaguchi, 756-0054 Japan
| | | | - Masanori Noda
- U-Medico Inc., 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masato Kiyoshi
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan
| | - Akira Harazono
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan
| | - Tetsuo Torisu
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Susumu Uchiyama
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Akiko Ishii-Watabe
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan
| |
Collapse
|
24
|
Real-time coating thickness measurement and defect recognition of film coated tablets with machine vision and deep learning. Int J Pharm 2022; 623:121957. [PMID: 35760260 DOI: 10.1016/j.ijpharm.2022.121957] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 11/22/2022]
Abstract
This paper presents a system, where images acquired with a digital camera are coupled with image analysis and deep learning to identify and categorize film coating defects and to measure the film coating thickness of tablets. There were 5 different classes of defective tablets, and the YOLOv5 algorithm was utilized to recognize defects, the accuracy of the classification was 98.2%. In order to characterize coating thickness, the diameter of the tablets in pixels was measured, which was used to measure the coating thickness of the tablets. The proposed system can be easily scaled up to match the production capability of continuous film coaters. With the developed technique, the complete screening of the produced tablets can be achieved in real-time resulting in the improvement of quality control.
Collapse
|
25
|
Tang K, Meyer Q, White R, Armstrong RT, Mostaghimi P, Da Wang Y, Liu S, Zhao C, Regenauer-Lieb K, Tung PKM. Deep learning for full-feature X-ray microcomputed tomography segmentation of proton electron membrane fuel cells. Comput Chem Eng 2022. [DOI: 10.1016/j.compchemeng.2022.107768] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
26
|
Alexandra Mészáros L, Farkas A, Madarász L, Bicsár R, László Galata D, Nagy B, Kristóf Nagy Z. UV/VIS imaging-based PAT tool for drug particle size inspection in intact tablets supported by pattern recognition neural networks. Int J Pharm 2022; 620:121773. [PMID: 35487400 DOI: 10.1016/j.ijpharm.2022.121773] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/09/2022] [Accepted: 04/22/2022] [Indexed: 11/26/2022]
Abstract
The potential of machine vision systems has not currently been exploited for pharmaceutical applications, although expected to provide revolutionary solutions for in-process and final product testing. The presented paper aimed to analyze the particle size of meloxicam, a yellow model active pharmaceutical ingredient, in intact tablets by a digital UV/VIS imaging-based machine vision system. Two image processing algorithms were developed and coupled with pattern recognition neural networks for UV and VIS images for particle size-based classification of the prepared tablets. The developed method can identify tablets containing finer or larger particles than the target with more than 97% accuracy. Two algorithms were developed for UV and VIS images for particle size analysis of the prepared tablets. According to the applied statistical tests, the obtained particle size distributions were similar to the results of the laser diffraction-based reference method. Digital UV/VIS imaging combined with multivariate data analysis can provide a new non-destructive, rapid, in-line tool for particle size analysis in tablets.
Collapse
Affiliation(s)
- Lilla Alexandra Mészáros
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, H-1111 Budapest, Műegyetem rakpart 3, Hungary
| | - Attila Farkas
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, H-1111 Budapest, Műegyetem rakpart 3, Hungary
| | - Lajos Madarász
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, H-1111 Budapest, Műegyetem rakpart 3, Hungary
| | - Rozália Bicsár
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, H-1111 Budapest, Műegyetem rakpart 3, Hungary
| | - Dorián László Galata
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, H-1111 Budapest, Műegyetem rakpart 3, Hungary
| | - Brigitta Nagy
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, H-1111 Budapest, Műegyetem rakpart 3, Hungary
| | - Zsombor Kristóf Nagy
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, H-1111 Budapest, Műegyetem rakpart 3, Hungary.
| |
Collapse
|
27
|
Puzzle Out Machine Learning Model-Explaining Disintegration Process in ODTs. Pharmaceutics 2022; 14:pharmaceutics14040859. [PMID: 35456693 PMCID: PMC9044744 DOI: 10.3390/pharmaceutics14040859] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/10/2022] [Accepted: 04/11/2022] [Indexed: 02/05/2023] Open
Abstract
Tablets are the most common dosage form of pharmaceutical products. While tablets represent the majority of marketed pharmaceutical products, there remain a significant number of patients who find it difficult to swallow conventional tablets. Such difficulties lead to reduced patient compliance. Orally disintegrating tablets (ODT), sometimes called oral dispersible tablets, are the dosage form of choice for patients with swallowing difficulties. ODTs are defined as a solid dosage form for rapid disintegration prior to swallowing. The disintegration time, therefore, is one of the most important and optimizable critical quality attributes (CQAs) for ODTs. Current strategies to optimize ODT disintegration times are based on a conventional trial-and-error method whereby a small number of samples are used as proxies for the compliance of whole batches. We present an alternative machine learning approach to optimize the disintegration time based on a wide variety of machine learning (ML) models through the H2O AutoML platform. ML models are presented with inputs from a database originally presented by Han et al., which was enhanced and curated to include chemical descriptors representing active pharmaceutical ingredient (API) characteristics. A deep learning model with a 10-fold cross-validation NRMSE of 8.1% and an R2 of 0.84 was obtained. The critical parameters influencing the disintegration of the directly compressed ODTs were ascertained using the SHAP method to explain ML model predictions. A reusable, open-source tool, the ODT calculator, is now available at Heroku platform.
Collapse
|
28
|
Floryanzia S, Ramesh P, Mills M, Kulkarni S, Chen G, Shah P, Lavrich D. Disintegration Testing Augmented by Computer Vision Technology. Int J Pharm 2022; 619:121668. [PMID: 35304245 DOI: 10.1016/j.ijpharm.2022.121668] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 01/10/2023]
Abstract
Oral solid dosage forms, specifically immediate release tablets, are prevalent in the pharmaceutical industry. Disintegration testing is often the first step of commercialization and large-scale production of these dosage forms. Current disintegration testing in the pharmaceutical industry, according to United States Pharmacopeia (USP) chapter <701>, only gives information about the duration of the tablet disintegration process. This information is subjective, variable, and prone to human error due to manual or physical data collection methods via the human eye or contact disks. To lessen the data integrity risk associated with this process, efforts have been made to automate the analysis of the disintegration process using digital lens and other imaging technologies. This would provide a non-invasive method to quantitatively determine disintegration time through computer algorithms. The main challenges associated with developing such a system involve visualization of tablet pieces through cloudy and turbid liquid. The Computer Vision for Disintegration (CVD) system has been developed to be used along with traditional pharmaceutical disintegration testing devices to monitor tablet pieces and distinguish them from the surrounding liquid. The software written for CVD utilizes data captured by cameras or other lenses then uses mobile SSD and CNN, with an OpenCV and FRCNN machine learning model, to analyze and interpreted the data. This technology is capable of consistently identifying tablets with ≥ 99.6% accuracy. Not only is the data produced by CVD more reliable, but it opens the possibility of a deeper understanding of disintegration rates and mechanisms in addition to duration.
Collapse
Affiliation(s)
- Sydney Floryanzia
- Merck & Co., Inc, 770 Sumneytown Pike, West Point, PA, USA; North Carolina State University, Raleigh, NC 27695
| | - Preethi Ramesh
- Merck & Co., Inc, 770 Sumneytown Pike, West Point, PA, USA; Syracuse University, Syracuse, NY 13244
| | - Madeline Mills
- Merck & Co., Inc, 770 Sumneytown Pike, West Point, PA, USA; Purdue University, West Lafayette, IN 47907
| | - Sanjana Kulkarni
- Merck & Co., Inc, 770 Sumneytown Pike, West Point, PA, USA; California Institute of Technology, Pasadena, CA 91125
| | - Grace Chen
- Merck & Co., Inc, 770 Sumneytown Pike, West Point, PA, USA.
| | - Prashant Shah
- Merck & Co., Inc, 770 Sumneytown Pike, West Point, PA, USA
| | - David Lavrich
- Merck & Co., Inc, 770 Sumneytown Pike, West Point, PA, USA
| |
Collapse
|
29
|
Deep Neural Networks to Detect Weeds from Crops in Agricultural Environments in Real-Time: A Review. REMOTE SENSING 2021. [DOI: 10.3390/rs13214486] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Automation, including machine learning technologies, are becoming increasingly crucial in agriculture to increase productivity. Machine vision is one of the most popular parts of machine learning and has been widely used where advanced automation and control have been required. The trend has shifted from classical image processing and machine learning techniques to modern artificial intelligence (AI) and deep learning (DL) methods. Based on large training datasets and pre-trained models, DL-based methods have proven to be more accurate than previous traditional techniques. Machine vision has wide applications in agriculture, including the detection of weeds and pests in crops. Variation in lighting conditions, failures to transfer learning, and object occlusion constitute key challenges in this domain. Recently, DL has gained much attention due to its advantages in object detection, classification, and feature extraction. DL algorithms can automatically extract information from large amounts of data used to model complex problems and is, therefore, suitable for detecting and classifying weeds and crops. We present a systematic review of AI-based systems to detect weeds, emphasizing recent trends in DL. Various DL methods are discussed to clarify their overall potential, usefulness, and performance. This study indicates that several limitations obstruct the widespread adoption of AI/DL in commercial applications. Recommendations for overcoming these challenges are summarized.
Collapse
|
30
|
The use of X-ray microtomography to investigate the microstructure of pharmaceutical tablets: Potentials and comparison to common physical methods. INTERNATIONAL JOURNAL OF PHARMACEUTICS-X 2021; 3:100090. [PMID: 34377974 PMCID: PMC8327351 DOI: 10.1016/j.ijpx.2021.100090] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 11/21/2022]
Abstract
Within this study, tablets microstructure was investigated by X-ray microtomgraphy. The aim was to gain information about their microstructure, and thus, derive deeper interpretation of tablet properties (mechanical strength, component distribution) and qualified property functions. Challenges in image processing are discussed for the correct identification of solids and voids. Furthermore, XMT measurements are critically compared with complementary physical methods for characterizing active pharmaceutical ingredient (API) content and porosity and its distribution (mercury porosimetry, calculated tablet porosity, Focused Ion Beam-Scanning Electron Microscopy (FIB-SEM)). The derived porosity by XMT is generally lower than the calculated porosity based on geometrical data due to the resolution of the XMT in relation to the pore sizes in tablets. With rising compactions stress and API concentration, deviations between the actual and the calculated API decrease. XMT showed that API clusters are present for all tablets containing >1 wt% of ibuprofen. The 3D orientation of the components is assessable by deriving cord lengths along all dimensions of the tablets. An increasing compaction stress leads to rising cord lengths, showing higher connectivity of the respective material. Its lesser extent in the z-direction illustrates the anisotropy of the compaction process. Additionally, cracks in the fabric are identified in tablets without visible macroscopic damage. Finally, the application of XMT provides valuable structural insights if its limitations are taken into account and its strengths are fostered by advanced pre- and post-processing.
Collapse
|
31
|
Kapoor Y, Meyer RF, Ferguson HM, Skomski D, Daublain P, Troup GM, Dalton C, Ramasamy M, Templeton AC. Flexibility in Drug Product Development: A Perspective. Mol Pharm 2021; 18:2455-2469. [PMID: 34165309 DOI: 10.1021/acs.molpharmaceut.1c00210] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The process of bringing a drug to market involves innumerable decisions to refine a concept into a final product. The final product goes through extensive research and development to meet the target product profile and to obtain a product that is manufacturable at scale. Historically, this process often feels inflexible and linear, as ideas and development paths are eliminated early on to allow focus on the workstream with the highest probability of success. Carrying multiple options early in development is both time-consuming and resource-intensive. Similarly, changing development pathways after significant investment carries a high "penalty of change" (PoC), which makes pivoting to a new concept late in development inhibitory. Can drug product (DP) development be made more flexible? The authors believe that combining a nonlinear DP development approach, leveraging state-of-the art data sciences, and using emerging process and measurement technologies will offer enhanced flexibility and should become the new normal. Through the use of iterative DP evaluation, "smart" clinical studies, artificial intelligence, novel characterization techniques, automation, and data collection/modeling/interpretation, it should be possible to significantly reduce the PoC during development. In this Perspective, a review of ideas/techniques along with supporting technologies that can be applied at each stage of DP development is shared. It is further discussed how these contribute to an improved and flexible DP development through the acceleration of the iterative build-measure-learn cycle in laboratories and clinical trials.
Collapse
Affiliation(s)
- Yash Kapoor
- Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Robert F Meyer
- Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Heidi M Ferguson
- Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Daniel Skomski
- Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Pierre Daublain
- Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Gregory M Troup
- Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Chad Dalton
- Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Manoharan Ramasamy
- Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Allen C Templeton
- Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| |
Collapse
|
32
|
Farkas D, Madarász L, Nagy ZK, Antal I, Kállai-Szabó N. Image Analysis: A Versatile Tool in the Manufacturing and Quality Control of Pharmaceutical Dosage Forms. Pharmaceutics 2021; 13:pharmaceutics13050685. [PMID: 34068724 PMCID: PMC8151645 DOI: 10.3390/pharmaceutics13050685] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 04/28/2021] [Accepted: 05/04/2021] [Indexed: 11/16/2022] Open
Abstract
In pharmaceutical sciences, visual inspection is one of the oldest methods used for description in pharmacopeias and is still an important part of the characterization and qualification of active ingredients, excipients, and dosage forms. With the development of technology, it is now also possible to take images of various pharmaceutical dosage forms with different imaging methods in a size range that is hardly visible or completely invisible to the human eye. By analyzing high-quality designs, physicochemical processes can be understood, and the results can be used even in the optimization of the composition of the dosage form and in the development of its production. The present study aims to show some of the countless ways image analysis can be used in the manufacturing and quality assessment of different dosage forms. This summary also includes measurements and an evaluation of, amongst others, a less studied dosage form, medicated foams.
Collapse
Affiliation(s)
- Dóra Farkas
- Department of Pharmaceutics, Semmelweis University, Hőgyes Str. 7, H-1092 Budapest, Hungary; (D.F.); (I.A.)
| | - Lajos Madarász
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, Műegyetem rakpart 3, H-1111 Budapest, Hungary; (L.M.); (Z.K.N.)
| | - Zsombor K. Nagy
- Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, Műegyetem rakpart 3, H-1111 Budapest, Hungary; (L.M.); (Z.K.N.)
| | - István Antal
- Department of Pharmaceutics, Semmelweis University, Hőgyes Str. 7, H-1092 Budapest, Hungary; (D.F.); (I.A.)
| | - Nikolett Kállai-Szabó
- Department of Pharmaceutics, Semmelweis University, Hőgyes Str. 7, H-1092 Budapest, Hungary; (D.F.); (I.A.)
- Correspondence:
| |
Collapse
|
33
|
Lou H, Lian B, Hageman MJ. Applications of Machine Learning in Solid Oral Dosage Form Development. J Pharm Sci 2021; 110:3150-3165. [PMID: 33951418 DOI: 10.1016/j.xphs.2021.04.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 02/07/2023]
Abstract
This review comprehensively summarizes the application of machine learning in solid oral dosage form development over the past three decades. In both academia and industry, machine learning is increasingly applied for multiple preformulation/formulation and process development studies. Further, this review provides the authors' perspectives on how pharmaceutical scientists can use machine learning for right projects and in right ways; some key ingredients include (1) the determination of inputs, outputs, and objectives; (2) the generation of a database containing high-quality data; (3) the development of machine learning models based on dataset training and model optimization; (4) the application of trained models in making predictions for new samples. It is expected by the authors and others that machine learning will promisingly play a more important role in tomorrow's projects for solid oral dosage form development.
Collapse
Affiliation(s)
- Hao Lou
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, United States; Biopharmaceutical Innovation and Optimization Center, University of Kansas, Lawrence, KS 66047, United States.
| | - Bo Lian
- College of Pharmacy, University of Arizona, Tucson, AZ 85721, United States
| | - Michael J Hageman
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, United States; Biopharmaceutical Innovation and Optimization Center, University of Kansas, Lawrence, KS 66047, United States
| |
Collapse
|
34
|
Paul S, Tseng YC. A semi-empirical model for estimation of flaw size in internally defective tablets. J Pharm Sci 2021; 110:2340-2345. [PMID: 33662393 DOI: 10.1016/j.xphs.2021.02.032] [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: 11/09/2020] [Revised: 01/27/2021] [Accepted: 02/24/2021] [Indexed: 11/28/2022]
Abstract
Capping is a mechanical defect in tablets, which is attributed to multiple factors including intrinsic material properties and tableting conditions. A suitable non-destructive approach using acoustically derived elastic modulus has showed distinctive features between a defective tablet and a defect-free tablet. In this work, a semi-empirical model was developed to estimate flaw size in an internally defective tablet from the relationship among elastic modulus, tablet density, and time of flight (acoustic wave to traverse through the tablet). The model was found fundamentally consistent where the derived flaw size showed clear dependence on powder mechanical properties of seven diverse formulations studied. Furthermore, the flaw size was reasonably correlated with the internal tablet microstructure illustrated by X-ray micro-tomography findings, both qualitatively and quantitatively. This model could thus be efficiently implemented for risk-based evaluation of internal defects in visibly intact tablets to ensure robustness of drug products.
Collapse
Affiliation(s)
- Shubhajit Paul
- Department of Material and Analytical Sciences, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06877, USA.
| | - Yin-Chao Tseng
- Department of Material and Analytical Sciences, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Road, Ridgefield, CT 06877, USA
| |
Collapse
|
35
|
Efficient Prediction of In Vitro Piroxicam Release and Diffusion From Topical Films Based on Biopolymers Using Deep Learning Models and Generative Adversarial Networks. J Pharm Sci 2021; 110:2531-2543. [PMID: 33548245 DOI: 10.1016/j.xphs.2021.01.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 12/12/2022]
Abstract
The purpose of this study was to simultaneously predict the drug release and skin permeation of Piroxicam (PX) topical films based on Chitosan (CTS), Xanthan gum (XG) and its Carboxymethyl derivatives (CMXs) as matrix systems. These films were prepared by the solvent casting method, using Tween 80 (T80) as a permeation enhancer. All of the prepared films were assessed for their physicochemical parameters, their in vitro drug release and ex vivo skin permeation studies. Moreover, deep learning models and machine learning models were applied to predict the drug release and permeation rates. The results indicated that all of the films exhibited good consistency and physicochemical properties. Furthermore, it was noticed that when T80 was used in the optimal formulation (F8) based on CTS-CMX3, a satisfactory drug release pattern was found where 99.97% of PX was released and an amount of 1.18 mg/cm2 was permeated after 48 h. Moreover, Generative Adversarial Network (GAN) efficiently enhanced the performance of deep learning models and DNN was chosen as the best predictive approach with MSE values equal to 0.00098 and 0.00182 for the drug release and permeation kinetics, respectively. DNN precisely predicted PX dissolution profiles with f2 values equal to 99.99 for all the formulations.
Collapse
|
36
|
Characterization of Spray Dried Particles Through Microstructural Imaging. J Pharm Sci 2020; 109:3404-3412. [DOI: 10.1016/j.xphs.2020.07.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/25/2020] [Accepted: 07/28/2020] [Indexed: 11/22/2022]
|