1
|
Wang M, Liu H, Huang J, Cai T, Xu ZP, Zhang L. Advancing cancer gene therapy: the emerging role of nanoparticle delivery systems. J Nanobiotechnology 2025; 23:362. [PMID: 40394591 PMCID: PMC12090605 DOI: 10.1186/s12951-025-03433-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 05/01/2025] [Indexed: 05/22/2025] Open
Abstract
Gene therapy holds immense potential due to its ability to precisely target oncogenes, making it a promising strategy for cancer treatment. Advances in genetic science and bioinformatics have expanded the applications of gene delivery technologies beyond detection and diagnosis to potential therapeutic interventions. However, traditional gene therapy faces significant challenges, including limited therapeutic efficacy and the rapid degradation of genetic materials in vivo. To address these limitations, multifunctional nanoparticles have been engineered to encapsulate and protect genetic materials, enhancing their stability and therapeutic effectiveness. Nanoparticles are being extensively explored for their ability to deliver various genetic payloads-including plasmid DNA, messenger RNA, and small interfering RNA-directly to cancer cells. This review highlights key gene modulation strategies such as RNA interference, gene editing systems, and chimeric antigen receptor (CAR) technologies, alongside a diverse array of nanoscale delivery systems composed of polymers, lipids, and inorganic materials. These nanoparticle-based delivery platforms aim to improve targeted transport of genetic material into cancer cells, ultimately enhancing the efficacy of cancer therapies.
Collapse
Affiliation(s)
- Maoze Wang
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, 315040, China
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518107, China
| | - Huina Liu
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, 315040, China
| | - Jinling Huang
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518107, China
- School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Ting Cai
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, 315040, China.
| | - Zhi Ping Xu
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, 315040, China.
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518107, China.
- School of Medicine, Hangzhou City University, Hangzhou, 310015, China.
| | - Lingxiao Zhang
- Interdisciplinary Nanoscience Center (INANO), Aarhus University, Aarhus C, DK-8000, Denmark.
| |
Collapse
|
2
|
Ferrari F, Berger J, Lemieux L, Paduraru C, Dillon M, Liaw A, Carrillo R, Wong S, Salami H, Avalle P, Sherer E, Richardson D, Skomski D. Bayesian hierarchical model predicts biopharmaceutical stability indicators and shelf life with application to multivalent human papillomavirus vaccine. Sci Rep 2025; 15:17333. [PMID: 40389588 PMCID: PMC12089594 DOI: 10.1038/s41598-025-99458-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 04/21/2025] [Indexed: 05/21/2025] Open
Abstract
Predictive stability is demonstrated as a powerful method for assessing the shelf-life of biopharmaceutical products, such as therapeutic proteins and vaccines. A Bayesian hierarchical multi-level stability model is illustrated for the Human Papillomavirus (HPV) 9-valent recombinant sub-unit vaccine GARDASIL®9. Ensuring speedy manufacturing and ample supply to satisfy the need of patients globally is pivotal, particularly for expanding vaccine access to underserved populations. Product heat stability and cold-chain supply play a major role in deployment of vaccines particularly to lower income countries, while lengthy real-time stability and shelf-life supporting studies are resource-intensive and time-consuming. Hence, an accelerated model-informed stability approach is devised. The product in this case study contains 9 molecular types (antigens) which each target different viral genotypes of the same class of the virus. The molecular types are mixed together as a co-formulation within a container (vial or syringe). The stability behavior of the platform vaccine was well-characterized experimentally and a single stability-limiting attribute was identified (potency). A Bayesian hierarchical stability model was developed utilizing long-term drug product storage data through shelf life at 5 °C as well as shorter-term accelerated stability data at 25 °C and 37 °C for 30 product batches. The model was able to comprehensively assess the stability of all molecular types in the vaccine as well as covariates like container type within a singular unified model framework. Moreover, method superiority was demonstrated for this application over multiple approaches such as linear and mixed effects models. This work elucidates that biopharmaceutical product platform knowledge from previous lots of a biopharmaceutical product in conjunction with batch-specific data from early stability timepoints can be employed to support long-term assessment for shelf-life of the stability and shelf-life indicating attribute(s). These findings, applied to two types vaccines including a multivalent vaccine, hold utility towards enabling accelerated patient access of future complex vaccines and biotherapeutic modalities. The results provide a novel framework for estimating a model for stability data in the context of evolving regulatory guidance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Paolo Avalle
- MSD Werthenstein BioPharma GmbH, Lucerne, Switzerland
| | | | | | | |
Collapse
|
3
|
Vidiyala N, Sunkishala P, Parupathi P, Nyavanandi D. The Role of Artificial Intelligence in Drug Discovery and Pharmaceutical Development: A Paradigm Shift in the History of Pharmaceutical Industries. AAPS PharmSciTech 2025; 26:133. [PMID: 40360908 DOI: 10.1208/s12249-025-03134-3] [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: 02/24/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
In today's world, with an increasing patient population, the need for medications is increasing rapidly. However, the current practice of drug development is time-consuming and requires a lot of investment by the pharmaceutical industries. Currently, it takes around 8-10 years and $3 billion of investment to develop a medication. Pharmaceutical industries and regulatory authorities are continuing to adopt new technologies to improve the efficiency of the drug development process. However, over the decades the pharmaceutical industries were not able to accelerate the drug development process. The pandemic (COVID-19) has taught the pharmaceutical industries and regulatory agencies an expensive lesson showing the need for emergency preparedness by accelerating the drug development process. Over the last few years, the pharmaceutical industries have been collaborating with artificial intelligence (AI) companies to develop algorithms and models that can be implemented at various stages of the drug development process to improve efficiency and reduce the developmental timelines significantly. In recent years, AI-screened drug candidates have entered clinical testing in human subjects which shows the interest of pharmaceutical companies and regulatory agencies. End-end integration of AI within the drug development process will benefit the industries for predicting the pharmacokinetic and pharmacodynamic profiles, toxicity, acceleration of clinical trials, study design, virtual monitoring of subjects, optimization of manufacturing process, analyzing and real-time monitoring of product quality, and regulatory preparedness. This review article discusses in detail the role of AI in various avenues of the pharmaceutical drug development process, its limitations, regulatory and future perspectives.
Collapse
Affiliation(s)
- Nithin Vidiyala
- Small Molecule Drug Product Development, Cerevel Therapeutics, Cambridge, Massachusetts, 02141, USA
| | - Pavani Sunkishala
- Process Validation, PCI Pharma Services, Bedford, New Hampshire, 03110, USA
| | - Prashanth Parupathi
- Division of Pharmaceutical Sciences, Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, New York, 11201, USA
| | - Dinesh Nyavanandi
- Small Molecule Drug Product Development, Cerevel Therapeutics, Cambridge, Massachusetts, 02141, USA.
| |
Collapse
|
4
|
Tarek M, El-Gogary RI, Kamel AO. A new era of psoriasis treatment: Drug repurposing through the lens of nanotechnology and machine learning. Int J Pharm 2025; 673:125385. [PMID: 39999900 DOI: 10.1016/j.ijpharm.2025.125385] [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: 12/24/2024] [Revised: 02/09/2025] [Accepted: 02/21/2025] [Indexed: 02/27/2025]
Abstract
Psoriasis is a persistent inflammatory skin disorder characterized by hyper-proliferation and abnormal epidermal differentiation. Conventional treatments such as; topical therapies, phototherapy, systemic immune modulators, and biologics aim to relieve symptoms and improve patient quality of life. However, challenges like adverse effects, high costs, and individual response variability persist. Thus, the need for novel anti-psoriatic drugs has led to the exploration of drug repurposing, an approach that identifies new applications for existing drugs. This method is in its early stages but has gained popularity across both public and private sectors. Furthermore, artificial intelligence (AI) integration is revolutionizing the healthcare industry by enhancing efficiency, delivery, and personalization. Machine learning and deep learning algorithms have significantly impacted drug discovery, repurposing, and designing new molecules or drug delivery carriers. Nanotechnology, in addition to AI, plays a pivotal role in targeting repurposed drugs via the topical route with suitable nanocarriers. This method overcomes challenges associated with oral delivery, such as systemic toxicities, slow onset of action, first-pass effect, and poor bioavailability. This review addresses the practice of repurposing existing drugs for managing psoriasis, discussing the challenges of conventional therapy and how the incorporation of nanotechnology and AI can overcome these hurdles, facilitating the discovery of anti-psoriatic drugs and presenting promising strategies for novel therapeutics. Additionally, it discusses the general benefits of drug repurposing compared to de novo drug development and the potential drawbacks of drug repurposing.
Collapse
Affiliation(s)
- Mahmoud Tarek
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt; Department of Pharmaceutics, Faculty of Pharmacy, Sinai University, Alarish, North Sinai 45511, Egypt
| | - Riham I El-Gogary
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Amany O Kamel
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt.
| |
Collapse
|
5
|
Rui M, Su Y, Tang H, Li Y, Fang N, Ge Y, Feng Q, Feng C. Computational Design and Optimization of Multi-Compound Multivesicular Liposomes for Co-Delivery of Traditional Chinese Medicine Compounds. AAPS PharmSciTech 2025; 26:61. [PMID: 39934607 DOI: 10.1208/s12249-025-03042-6] [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: 08/19/2024] [Accepted: 01/08/2025] [Indexed: 02/13/2025] Open
Abstract
Study explored the synergistic anti-tumor effects of a combination of compounds from Traditional Chinese Medicine, including rosmarinic acid (RA), chlorogenic acid (CA), and scoparone (SCO), in the formulation of multivesicular liposomes (MVLs). Optimization of formulations and process parameters was essential to achieve effective liposomal encapsulation and optimal release profiles for these three compounds with diverse properties. Traditional trial-and-error approaches are inefficient for the optimization of complex multi-compound MVLs. We developed a new formulation optimization model, which could address this issue by predicting the optimal multi-compound MVLs formulation. Our machine learning model integrated support vector machine regression (SVR) algorithm and cuckoo search (CS) algorithm, resulting in three CS-SVR models to predict single-compound MVLs. The CS algorithm, with various weighting rules, was then applied to search the best formulation parameters across three CS-SVR models and to maximize the encapsulation efficiency for all three compounds. The multi-compound MLVs were subsequently prepared under the predicted conditions, achieving an optimized particle size of 15.12 µm, with encapsulation efficiencies of 82.93 ± 2.43% for CA, 82.22 ± 1.25% for RA, and 95.60 ± 0.18% for SCO. The predicted optimal multi-compound MVLs were further validated through in vitro characterization and in vivo anti-tumor experiments, showing a promising synergistic anti-tumor effect consistent with in vitro results. This model accurately predicted optimal encapsulation conditions, which were validated experimentally, demonstrating improved encapsulation efficiencies and reduced trial-and-error iterations. Collectively, our model provides a predictive pathway for multi-compound MVLs formulation, indicating the ability of this model to significantly reduce experimental burden and accelerate formulation development.
Collapse
Affiliation(s)
- Mengjie Rui
- School of Pharmacy, Jiangsu University, No.301 Xuefu Road, Zhenjiang, 212013, Jiangsu Province, China
| | - Yali Su
- School of Pharmacy, Jiangsu University, No.301 Xuefu Road, Zhenjiang, 212013, Jiangsu Province, China
| | - Haidan Tang
- School of Pharmacy, Jiangsu University, No.301 Xuefu Road, Zhenjiang, 212013, Jiangsu Province, China
| | - Yinfeng Li
- School of Pharmacy, Jiangsu University, No.301 Xuefu Road, Zhenjiang, 212013, Jiangsu Province, China
| | - Naying Fang
- School of Pharmacy, Jiangsu University, No.301 Xuefu Road, Zhenjiang, 212013, Jiangsu Province, China
| | - Yingying Ge
- School of Pharmacy, Jiangsu University, No.301 Xuefu Road, Zhenjiang, 212013, Jiangsu Province, China
| | - Qiuqi Feng
- School of Pharmacy, Jiangsu University, No.301 Xuefu Road, Zhenjiang, 212013, Jiangsu Province, China
| | - Chunlai Feng
- Department of Obstetrics, Affiliated Hospital of Jiangsu University, No.438 Jiefang Road, Zhenjiang, 212001, Jiangsu Province, China.
- School of Pharmacy, Jiangsu University, No.301 Xuefu Road, Zhenjiang, 212013, Jiangsu Province, China.
| |
Collapse
|
6
|
Wang N, Dong J, Ouyang D. AI-directed formulation strategy design initiates rational drug development. J Control Release 2025; 378:619-636. [PMID: 39719215 DOI: 10.1016/j.jconrel.2024.12.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/27/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024]
Abstract
Rational drug development would be impossible without selecting the appropriate formulation route. However, pharmaceutical scientists often rely on limited personal experiences to perform trial-and-error tests on diverse formulation strategies. Such an inefficient screening manner not only wastes research investments but also threatens the safety of clinical volunteers and patients. A design-oriented paradigm for formulation strategy determination is urgently needed to initiate rational drug development. Herein, we introduce FormulationDT, the first data-driven and knowledge-guided artificial intelligence (AI) platform for rational formulation strategy design. Learning from approved drug formulations, FormulationDT devised a comprehensive formulation strategy design system containing 12 decisions for both oral and injectable administration. Utilizing PU-Decide, our specialized partially supervised learning framework designed for positive-unlabeled (PU) scenarios, FormulationDT developed precise and interpretable classification models for each decision, achieving area under the receiver operating characteristic curve (ROC_AUC) scores ranging from 0.78 to 0.98, with an average above 0.90. Incorporating extensive domain knowledge, FormulationDT is now accessible through a user-friendly web platform (http://formulationdt.computpharm.org/). Moreover, FormulationDT demonstrates its value by showcasing its application in proteolysis targeting chimeras (PROTACs) and recent drug approvals. Overall, this study created the first approved drug formulation dataset and tailored the PU-Decide framework to develop a high-performance, interpretable, and user-friendly AI formulation strategy design platform, which holds promise for driving risk reduction and efficiency gains across the life cycle of drug discovery and development.
Collapse
Affiliation(s)
- Nannan Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, Macau, China
| | - Jie Dong
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.
| | - Defang Ouyang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, Macau, China; Department of Public Health and Medicinal Administration, Faculty of Health Sciences (FHS), University of Macau, Macau, China.
| |
Collapse
|
7
|
Borja M, Dhondt J, Bertels J, Van Hauwermeiren D, Verwaeren J. Modelling the effect of base component properties and processing conditions on mixture products using probabilistic, knowledge-guided neural networks. Int J Pharm 2025; 669:125012. [PMID: 39643149 DOI: 10.1016/j.ijpharm.2024.125012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/09/2024]
Abstract
Development of materials by mixing different base components is a widespread methodology to create materials with improved properties compared to those of its base components. However, efficient determination of the properties of mixture-based materials during design remains challenging without prior knowledge of the underlying physical phenomena. In this work a new data-based methodology is proposed involving the use of probabilistic, knowledge-guided artificial neural networks to jointly model the properties of the base components, the proportions in which they are mixed, and the processing conditions used during manufacture to predict properties of final products. The method proposed does not involve any assumptions in terms of ideal mixing rules of the base components, and allows for estimation of aleatoric uncertainty in the prediction. Additionally, an extension is presented that incorporates expert knowledge into the model by the implementation of monotonicity constraints between certain inputs and outputs. The methodology is illustrated with a case study involving the formulation of drug products using direct compression. The model is used to predict pharmaceutical tablets' quality attributes (mass variation, tensile strength, disintegration time, friability and ejection force), showing that the method is able to predict properties of the final product overcoming gaps currently present in previous modelling approaches.
Collapse
Affiliation(s)
- Manuel Borja
- Department of Data Analysis and Mathematical Modelling, Ghent University, Coupure links 653, Gent, 9000, Belgium.
| | - Jens Dhondt
- Oral Solids Development, Pharmaceutical Product Development & Supply, Pharmaceutical Research and Development, Division of Janssen Pharmaceutica, Johnson & Johnson, Turnhoutseweg 30, Beerse, B-2340, Belgium
| | - Johny Bertels
- Oral Solids Development, Pharmaceutical Product Development & Supply, Pharmaceutical Research and Development, Division of Janssen Pharmaceutica, Johnson & Johnson, Turnhoutseweg 30, Beerse, B-2340, Belgium
| | - Daan Van Hauwermeiren
- Department of Data Analysis and Mathematical Modelling, Ghent University, Coupure links 653, Gent, 9000, Belgium
| | - Jan Verwaeren
- Department of Data Analysis and Mathematical Modelling, Ghent University, Coupure links 653, Gent, 9000, Belgium
| |
Collapse
|
8
|
Dhaval M, Dudhat K, Gadoya A, Shah S, Pethani T, Jambukiya N, Patel A, Kalsariya C, Ansari J, Borkhataria C. Pharmaceutical Salts: Comprehensive Insights From Fundamental Chemistry to FDA Approvals (2019-2023). AAPS PharmSciTech 2025; 26:36. [PMID: 39821716 DOI: 10.1208/s12249-024-03020-4] [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: 08/13/2024] [Accepted: 12/05/2024] [Indexed: 01/19/2025] Open
Abstract
Pharmaceutical salts are a cornerstone in drug development, offering a robust, economical, and industry-friendly option for improving the crucial physicochemical properties of drugs, particularly solubility and dissolution. This review article explores all critical aspects of salt formation, including its importance, the basic chemistry involved, the principles governing counterion selection, the range of counterions used, and the methods for preparing salts along with their advantages and limitations. Additionally, it explores analytical techniques for confirming salt formation and the different approaches various countries adopt in considering new salts as intellectual property. Furthermore, the review sheds light on US FDA-approved salts from 2019 to 2023, providing a unique perspective by analyzing trends in counterion selection observed in FDA-approved salts during this period. Despite the extensive literature on pharmaceutical salts, a comprehensive review addressing all these critical aspects in a single article with a focus on current trends and particularly on US FDA-approved salts from 2019 to 2023 is lacking. This review bridges this gap by thoroughly exploring all mentioned facets of pharmaceutical salts and providing an up-to-date overview.
Collapse
Affiliation(s)
- Mori Dhaval
- B.K. Mody Government Pharmacy College, Polytechnic Campus, Near Ajidam, Rajkot, Gujarat-360005, India.
| | - Kiran Dudhat
- R.K. School of Pharmacy, R.K. University, Rajkot, Gujarat, India
| | - Aastha Gadoya
- B.K. Mody Government Pharmacy College, Polytechnic Campus, Near Ajidam, Rajkot, Gujarat-360005, India
| | - Sunny Shah
- B.K. Mody Government Pharmacy College, Polytechnic Campus, Near Ajidam, Rajkot, Gujarat-360005, India
| | - Trupesh Pethani
- Department of Pharmaceutical Sciences, Saurashtra University, Rajkot, Gujarat, India
| | - Nilesh Jambukiya
- B.K. Mody Government Pharmacy College, Polytechnic Campus, Near Ajidam, Rajkot, Gujarat-360005, India
| | - Ajay Patel
- B.K. Mody Government Pharmacy College, Polytechnic Campus, Near Ajidam, Rajkot, Gujarat-360005, India
| | - Chintan Kalsariya
- B.K. Mody Government Pharmacy College, Polytechnic Campus, Near Ajidam, Rajkot, Gujarat-360005, India
| | - Jainabparvin Ansari
- B.K. Mody Government Pharmacy College, Polytechnic Campus, Near Ajidam, Rajkot, Gujarat-360005, India
| | - Chetan Borkhataria
- B.K. Mody Government Pharmacy College, Polytechnic Campus, Near Ajidam, Rajkot, Gujarat-360005, India
| |
Collapse
|
9
|
Zulbeari N, Wang F, Mustafova SS, Parhizkar M, Holm R. Machine learning strengthened formulation design of pharmaceutical suspensions. Int J Pharm 2025; 668:124967. [PMID: 39566699 DOI: 10.1016/j.ijpharm.2024.124967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 11/22/2024]
Abstract
Many different formulation strategies have been investigated to oppose suboptimal treatment of long-term or chronic conditions, one of which are the nano- and microsuspensions prepared as long-acting injectables to prolong the release of an active pharmaceutical compound for a defined period of time by regulating the size of particles by milling. Typically, surfactant and/or polymers are added in the dispersion medium of the suspension during processing for stabilization purposes. However, current formulation investigations with milling are heavily based on prior expertise and trial-and-error approaches. Various interacting parameters such as the milling bead size, stabilizer type and concentration have confounded the investigation of milling process. The present study systematically exploited statistical and machine learning (ML) strategies to understand the relationship between suspension characteristics and formulation parameters under full-factorial milling experiments. Stabilizer concentration was identified as a significant factor (p < 0.001) for median suspension diameter (D50). A formulation stability classification ML model with high prediction accuracy (0.91) and F1-score (0.91) under 10-fold cross-validation was constructed based on 72 formulation datapoints. Model interpretation through Shapley additive explanations (SHAP) revealed the prominent impact of stabilizer concentration and milling bead size on formulation stability. The present work demonstrated the potential to achieve a deeper understanding of the design and optimization of nano- and microsuspensions through explainable ML modelling on formulation screening data.
Collapse
Affiliation(s)
- Nadina Zulbeari
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Fanjin Wang
- Deparment of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, WC1N 1AX London, United Kingdom
| | - Sibel Selyatinova Mustafova
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Maryam Parhizkar
- Deparment of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, WC1N 1AX London, United Kingdom
| | - René Holm
- Department of Physics, Chemistry, and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark.
| |
Collapse
|
10
|
Matalqah S, Lafi Z, Mhaidat Q, Asha N, Yousef Asha S. 'Applications of machine learning in liposomal formulation and development'. Pharm Dev Technol 2025; 30:126-136. [PMID: 39780760 DOI: 10.1080/10837450.2024.2448777] [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: 10/12/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025]
Abstract
Machine learning (ML) has emerged as a transformative tool in drug delivery, particularly in the design and optimization of liposomal formulations. This review focuses on the intersection of ML and liposomal technology, highlighting how advanced algorithms are accelerating formulation processes, predicting key parameters, and enabling personalized therapies. ML-driven approaches are restructuring formulation development by optimizing liposome size, stability, and encapsulation efficiency while refining drug release profiles. Additionally, the integration of ML enhances therapeutic outcomes by enabling precision-targeted delivery and minimizing side effects. This review presents current breakthroughs, challenges, and future opportunities in applying ML to liposomal systems, aiming to improve therapeutic efficacy and patient outcomes in various disease treatments.
Collapse
Affiliation(s)
- Sina Matalqah
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Zainab Lafi
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | | | | | | |
Collapse
|
11
|
Wang W, Chen K, Jiang T, Wu Y, Wu Z, Ying H, Yu H, Lu J, Lin J, Ouyang D. Artificial intelligence-driven rational design of ionizable lipids for mRNA delivery. Nat Commun 2024; 15:10804. [PMID: 39738043 PMCID: PMC11685617 DOI: 10.1038/s41467-024-55072-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 11/29/2024] [Indexed: 01/01/2025] Open
Abstract
Lipid nanoparticles (LNPs) have proven effective in mRNA delivery, as evidenced by COVID-19 vaccines. Its key ingredient, ionizable lipids, is traditionally optimized by inefficient and costly experimental screening. This study leverages artificial intelligence (AI) and virtual screening to facilitate the rational design of ionizable lipids by predicting two key properties of LNPs, apparent pKa and mRNA delivery efficiency. Nearly 20 million ionizable lipids were evaluated through two iterations of AI-driven generation and screening, yielding three and six new molecules, respectively. In mouse test validation, one lipid from the initial iteration, featuring a benzene ring, demonstrated performance comparable to the control DLin-MC3-DMA (MC3). Notably, all six lipids from the second iteration equaled or outperformed MC3, with one exhibiting efficacy akin to a superior control lipid SM-102. Furthermore, the AI model is interpretable in structure-activity relationships.
Collapse
Affiliation(s)
- Wei Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Kepan Chen
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
- Center for mRNA Translational Research, Fudan University, Shanghai, China
| | - Ting Jiang
- Center for mRNA Translational Research, Fudan University, Shanghai, China
- Shanghai RNACure Biopharma Co., Ltd, Shanghai, China
| | - Yiyang Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Zheng Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Hang Ying
- Center for mRNA Translational Research, Fudan University, Shanghai, China
- Shanghai RNACure Biopharma Co., Ltd, Shanghai, China
| | - Hang Yu
- Center for mRNA Translational Research, Fudan University, Shanghai, China
- Shanghai RNACure Biopharma Co., Ltd, Shanghai, China
| | - Jing Lu
- Center for mRNA Translational Research, Fudan University, Shanghai, China
- Shanghai RNACure Biopharma Co., Ltd, Shanghai, China
| | - Jinzhong Lin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China.
- Center for mRNA Translational Research, Fudan University, Shanghai, China.
| | - Defang Ouyang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China.
- Faculty of Health Sciences, University of Macau, Macau, China.
| |
Collapse
|
12
|
Pouyanfar N, Anvari Z, Davarikia K, Aftabi P, Tajik N, Shoara Y, Ahmadi M, Ayyoubzadeh SM, Shahbazi MA, Ghorbani-Bidkorpeh F. Machine learning-assisted rheumatoid arthritis formulations: A review on smart pharmaceutical design. MATERIALS TODAY COMMUNICATIONS 2024; 41:110208. [DOI: 10.1016/j.mtcomm.2024.110208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
13
|
Dorsey PJ, Lau CL, Chang TC, Doerschuk PC, D'Addio SM. Review of machine learning for lipid nanoparticle formulation and process development. J Pharm Sci 2024; 113:3413-3433. [PMID: 39341497 DOI: 10.1016/j.xphs.2024.09.015] [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: 06/08/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 10/01/2024]
Abstract
Lipid nanoparticles (LNPs) are a subset of pharmaceutical nanoparticulate formulations designed to encapsulate, stabilize, and deliver nucleic acid cargoes in vivo. Applications for LNPs include new interventions for genetic disorders, novel classes of vaccines, and alternate modes of intracellular delivery for therapeutic proteins. In the pharmaceutical industry, establishing a robust formulation and process to achieve target product performance is a critical component of drug development. Fundamental understanding of the processes for making LNPs and their interactions with biological systems have advanced considerably in the wake of the COVID-19 pandemic. Nevertheless, LNP formulation research remains largely empirical and resource intensive due to the multitude of input parameters and the complex physical phenomena that govern the processes of nanoparticle precipitation, self-assembly, structure evolution, and stability. Increasingly, artificial intelligence and machine learning (AI/ML) are being applied to improve the efficiency of research activities through in silico models and predictions, and to drive deeper fundamental understanding of experimental inputs to functional outputs. This review will identify current challenges and opportunities in the development of robust LNP formulations of nucleic acids, review studies that apply machine learning methods to experimental datasets, and provide discussion on associated data science challenges to facilitate collaboration between formulation and data scientists, aiming to accelerate the advancement of AI/ML applied to LNP formulation and process optimization.
Collapse
Affiliation(s)
- Phillip J Dorsey
- Pharmaceutical Sciences & Clinical Supply, MRL, Merck & Co., Inc., Rahway, NJ 07065, USA; University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Christina L Lau
- Cornell University, School of Electrical and Computer Engineering, Ithaca, NY 14853, USA
| | - Ti-Chiun Chang
- Pharmaceutical Sciences & Clinical Supply, MRL, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Peter C Doerschuk
- Cornell University, School of Electrical and Computer Engineering, Ithaca, NY 14853, USA
| | - Suzanne M D'Addio
- Pharmaceutical Sciences & Clinical Supply, MRL, Merck & Co., Inc., Rahway, NJ 07065, USA.
| |
Collapse
|
14
|
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
|
15
|
Sousa AS, Serra J, Estevens C, Costa R, Ribeiro AJ. Unveiling Swelling and Erosion Dynamics: Early Development Screening of Mirabegron Extended Release Tablets. AAPS PharmSciTech 2024; 25:277. [PMID: 39604660 DOI: 10.1208/s12249-024-02994-5] [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: 08/22/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Although the development of extended release (ER) matrices has been extensively investigated, understanding the most appropriate mechanism of drug release to achieve the desired release remains a cost- and time-consuming challenge in the early stages of formulation development. This study aimed to investigate the early stage of developing ER hydrophilic matrix tablets containing mirabegron as a model drug, focusing on the effects of polymer type, diluent type, and polymer amount on critical quality attributes (CQAs), namely, tablet swelling and erosion behavior. A full factorial design was employed to explore the interactions of control factors through multivariate regression analysis, emphasizing the application of quality by design (QbD) principles. The swelling and erosion performances of 72 formulations were evaluated. The swelling data were fitted to the Vergnaud model. Finally, in vitro drug release profiles were investigated for four of the formulations studied. The polymer type, diluent type, and polymer amount had distinct effects on the swelling and erosion behavior of the ER matrix tablets. Compared with those with isomalt (G720) or dextrate (DXT), formulations with polyethylene glycol 8000 (P8000) consistently exhibited greater swelling. Additionally, higher molecular weight was correlated with increased swelling within the same polymer type. Hydroxypropylmethylcellulose (HPMC) and polyethylene oxide (PEO)-based formulations showed higher swelling rates, while polyvinyl alcohol (PVA-80) displayed the highest erosion percentage. The findings highlight the significance of incorporating early-stage screening designs to maximize efficiency and optimize time and resource. This approach enables the development of a comprehensive understanding of drug release mechanisms from ER matrix tablets.
Collapse
Affiliation(s)
- Ana S Sousa
- Faculdade de Farmácia, Universidade de Coimbra, Coimbra, 3000-148, Portugal
- Grupo Tecnimede, Quinta da Cerca, Caixaria, Dois Portos, 2565-187, Portugal
| | - J Serra
- Grupo Tecnimede, Quinta da Cerca, Caixaria, Dois Portos, 2565-187, Portugal
| | - C Estevens
- Grupo Tecnimede, Quinta da Cerca, Caixaria, Dois Portos, 2565-187, Portugal
| | - R Costa
- Grupo Tecnimede, Quinta da Cerca, Caixaria, Dois Portos, 2565-187, Portugal
| | - António J Ribeiro
- Faculdade de Farmácia, Universidade de Coimbra, Coimbra, 3000-148, Portugal.
- i3S, IBMC, Porto, Rua Alfredo Allen, 4200-135, Portugal.
| |
Collapse
|
16
|
Yang L, Cao S, Liu L, Zhu R, Wu D. cyclicpeptide: a Python package for cyclic peptide drug design. Brief Bioinform 2024; 26:bbae714. [PMID: 39783893 PMCID: PMC11713021 DOI: 10.1093/bib/bbae714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/12/2024] [Accepted: 12/27/2024] [Indexed: 01/12/2025] Open
Abstract
The unique cyclic structure of cyclic peptides grants them remarkable stability and bioactivity, making them powerful candidates for treating various diseases. However, the lack of standardized tools for cyclic peptide data has hindered their potential in today's artificial intelligence-driven efficient drug design landscape. To bridge this gap, here we introduce a Python package named cyclicpeptide specifically for cyclic peptide drug design. This package provides standardized tools such as Structure2Sequence, Sequence2Structure, and format transformation to process, convert, and standardize cyclic peptide structure and sequence data. Additionally, it includes GraphAlignment for cyclic peptide-specific alignment and search and PropertyAnalysis to enhance the understanding of their drug-like properties and potential applications. This comprehensive suite of tools aims to streamline the integration of cyclic peptides into modern drug discovery pipelines, accelerating the development of cyclic peptide-based therapeutics.
Collapse
Affiliation(s)
- Liu Yang
- National Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou 310052, P. R. China
| | - Suqi Cao
- National Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou 310052, P. R. China
| | - Lei Liu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200072, P. R. China
| | - Ruixin Zhu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200072, P. R. China
| | - Dingfeng Wu
- National Center, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou 310052, P. R. China
| |
Collapse
|
17
|
Xie B, Liu Y, Li X, Yang P, He W. Solubilization techniques used for poorly water-soluble drugs. Acta Pharm Sin B 2024; 14:4683-4716. [PMID: 39664427 PMCID: PMC11628819 DOI: 10.1016/j.apsb.2024.08.027] [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: 05/18/2024] [Revised: 07/28/2024] [Accepted: 08/14/2024] [Indexed: 12/13/2024] Open
Abstract
About 40% of approved drugs and nearly 90% of drug candidates are poorly water-soluble drugs. Low solubility reduces the drugability. Effectively improving the solubility and bioavailability of poorly water-soluble drugs is a critical issue that needs to be urgently addressed in drug development and application. This review briefly introduces the conventional solubilization techniques such as solubilizers, hydrotropes, cosolvents, prodrugs, salt modification, micronization, cyclodextrin inclusion, solid dispersions, and details the crystallization strategies, ionic liquids, and polymer-based, lipid-based, and inorganic-based carriers in improving solubility and bioavailability. Some of the most commonly used approved carrier materials for solubilization techniques are presented. Several approved poorly water-soluble drugs using solubilization techniques are summarized. Furthermore, this review summarizes the solubilization mechanism of each solubilization technique, reviews the latest research advances and challenges, and evaluates the potential for clinical translation. This review could guide the selection of a solubilization approach, dosage form, and administration route for poorly water-soluble drugs. Moreover, we discuss several promising solubilization techniques attracting increasing attention worldwide.
Collapse
Affiliation(s)
- Bing Xie
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Yaping Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Pei Yang
- School of Science, China Pharmaceutical University, Nanjing 2111198, China
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| |
Collapse
|
18
|
Bannigan P, Hickman RJ, Aspuru‐Guzik A, Allen C. The Dawn of a New Pharmaceutical Epoch: Can AI and Robotics Reshape Drug Formulation? Adv Healthc Mater 2024; 13:e2401312. [PMID: 39155417 PMCID: PMC11582498 DOI: 10.1002/adhm.202401312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/21/2024] [Indexed: 08/20/2024]
Abstract
Over the last four decades, pharmaceutical companies' expenditures on research and development have increased 51-fold. During this same time, clinical success rates for new drugs have remained unchanged at about 10 percent, predominantly due to lack of efficacy and/or safety concerns. This persistent problem underscores the need to innovate across the entire drug development process, particularly in drug formulation, which is often deprioritized and under-resourced.
Collapse
Affiliation(s)
- Pauric Bannigan
- Intrepid Labs Inc.MaRS CentreWest Tower661 University Avenue Suite 1300TorontoONM5G 0B7Canada
| | - Riley J. Hickman
- Intrepid Labs Inc.MaRS CentreWest Tower661 University Avenue Suite 1300TorontoONM5G 0B7Canada
| | - Alán Aspuru‐Guzik
- Intrepid Labs Inc.MaRS CentreWest Tower661 University Avenue Suite 1300TorontoONM5G 0B7Canada
- Department of Chemical Engineering and Applied ChemistryUniversity of TorontoTorontoONM5S 3E5Canada
- Acceleration ConsortiumUniversity of TorontoTorontoONM5S 3H6Canada
- Department of ChemistryUniversity of TorontoTorontoONM5S 3H6Canada
| | - Christine Allen
- Intrepid Labs Inc.MaRS CentreWest Tower661 University Avenue Suite 1300TorontoONM5G 0B7Canada
- Department of Chemical Engineering and Applied ChemistryUniversity of TorontoTorontoONM5S 3E5Canada
- Acceleration ConsortiumUniversity of TorontoTorontoONM5S 3H6Canada
- Leslie Dan Faculty of PharmacyUniversity of TorontoTorontoONM5S 3M2Canada
| |
Collapse
|
19
|
Nkune NW, Abrahamse H. Possible integration of artificial intelligence with photodynamic therapy and diagnosis: A review. J Drug Deliv Sci Technol 2024; 101:106210. [DOI: 10.1016/j.jddst.2024.106210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
20
|
Xie X, Xiao YF, Yang H, Peng X, Li JJ, Zhou YY, Fan CQ, Meng RP, Huang BB, Liao XP, Chen YY, Zhong TT, Lin H, Koulaouzidis A, Yang SM. A new artificial intelligence system for both stomach and small-bowel capsule endoscopy. Gastrointest Endosc 2024; 100:878.e1-878.e14. [PMID: 38851456 DOI: 10.1016/j.gie.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 05/31/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND AND AIMS Despite the benefits of artificial intelligence in small-bowel (SB) capsule endoscopy (CE) image reading, information on its application in the stomach and SB CE is lacking. METHODS In this multicenter, retrospective diagnostic study, gastric imaging data were added to the deep learning-based SmartScan (SS), which has been described previously. A total of 1069 magnetically controlled GI CE examinations (comprising 2,672,542 gastric images) were used in the training phase for recognizing gastric pathologies, producing a new artificial intelligence algorithm named SS Plus. A total of 342 fully automated, magnetically controlled CE examinations were included in the validation phase. The performance of both senior and junior endoscopists with both the SS Plus-assisted reading (SSP-AR) and conventional reading (CR) modes was assessed. RESULTS SS Plus was designed to recognize 5 types of gastric lesions and 17 types of SB lesions. SS Plus reduced the number of CE images required for review to 873.90 (median, 1000; interquartile range [IQR], 814.50-1000) versus 44,322.73 (median, 42,393; IQR, 31,722.75-54,971.25) for CR. Furthermore, with SSP-AR, endoscopists took 9.54 minutes (median, 8.51; IQR, 6.05-13.13) to complete the CE video reading. In the 342 CE videos, SS Plus identified 411 gastric and 422 SB lesions, whereas 400 gastric and 368 intestinal lesions were detected with CR. Moreover, junior endoscopists remarkably improved their CE image reading ability with SSP-AR. CONCLUSIONS Our study shows that the newly upgraded deep learning-based algorithm SS Plus can detect GI lesions and help improve the diagnostic performance of junior endoscopists in interpreting CE videos.
Collapse
Affiliation(s)
- Xia Xie
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Yu-Feng Xiao
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Huan Yang
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Xue Peng
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Jian-Jun Li
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Yuan-Yuan Zhou
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Chao-Qiang Fan
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Rui-Ping Meng
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Bao-Bao Huang
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Xi-Ping Liao
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Yu-Yang Chen
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Ting-Ting Zhong
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Hui Lin
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China; Department of Epidemiology, the Third Military Medical University, Chongqing, China.
| | - Anastasios Koulaouzidis
- Department of Clinical Research University of Southern Denmark, Odense, Denmark; Centre for Clinical Implementation of Capsule Endoscopy, Store Adenomer Tidlige Cancere Centre, Svendborg, Denmark.
| | - Shi-Ming Yang
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China.
| |
Collapse
|
21
|
Bao Z, Tom G, Cheng A, Watchorn J, Aspuru-Guzik A, Allen C. Towards the prediction of drug solubility in binary solvent mixtures at various temperatures using machine learning. J Cheminform 2024; 16:117. [PMID: 39468626 PMCID: PMC11520512 DOI: 10.1186/s13321-024-00911-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/28/2024] [Indexed: 10/30/2024] Open
Abstract
Drug solubility is an important parameter in the drug development process, yet it is often tedious and challenging to measure, especially for expensive drugs or those available in small quantities. To alleviate these challenges, machine learning (ML) has been applied to predict drug solubility as an alternative approach. However, the majority of existing ML research has focused on the predictions of aqueous solubility and/or solubility at specific temperatures, which restricts the model applicability in pharmaceutical development. To bridge this gap, we compiled a dataset of 27,000 solubility datapoints, including solubility of small molecules measured in a range of binary solvent mixtures under various temperatures. Next, a panel of ML models were trained on this dataset with their hyperparameters tuned using Bayesian optimization. The resulting top-performing models, both gradient boosted decision trees (light gradient boosting machine and extreme gradient boosting), achieved mean absolute errors (MAE) of 0.33 for LogS (S in g/100 g) on the holdout set. These models were further validated through a prospective study, wherein the solubility of four drug molecules were predicted by the models and then validated with in-house solubility experiments. This prospective study demonstrated that the models accurately predicted the solubility of solutes in specific binary solvent mixtures under different temperatures, especially for drugs whose features closely align within the solutes in the dataset (MAE < 0.5 for LogS). To support future research and facilitate advancements in the field, we have made the dataset and code openly available. Scientific contribution Our research advances the state-of-the-art in predicting solubility for small molecules by leveraging ML and a uniquely comprehensive dataset. Unlike existing ML studies that predominantly focus on solubility in aqueous solvents at fixed temperatures, our work enables prediction of drug solubility in a variety of binary solvent mixtures over a broad temperature range, providing practical insights on the modeling of solubility for realistic pharmaceutical applications. These advancements along with the open access dataset and code support significant steps in the drug development process including new molecule discovery, drug analysis and formulation.
Collapse
Affiliation(s)
- Zeqing Bao
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Gary Tom
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, ON, M5S 1M1, Canada
| | - Austin Cheng
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, ON, M5S 1M1, Canada
| | | | - Alán Aspuru-Guzik
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, ON, M5S 1M1, Canada
- Acceleration Consortium, Toronto, ON, M5S 3H6, Canada
- Lebovic Fellow, Canadian Institute for Advanced Research (CIFAR), Toronto, ON, M5S 1M1, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, M5S 3E5, Canada
- Department of Materials Science and Engineering, University of Toronto, Toronto, ON, M5S 3E4, Canada
- CIFAR Artificial Intelligence Research Chair, Vector Institute, Toronto, ON, M5S 1M1, Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada.
- Acceleration Consortium, Toronto, ON, M5S 3H6, Canada.
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, M5S 3E5, Canada.
| |
Collapse
|
22
|
Zhao J, Hermans E, Sepassi K, Tistaert C, Bergström CAS, Ahmad M, Larsson P. Effect of Data Quality and Data Quantity on the Estimation of Intrinsic Solubility: Analysis Based on a Single-Source Data Set. Mol Pharm 2024; 21:5261-5271. [PMID: 39267585 PMCID: PMC11462503 DOI: 10.1021/acs.molpharmaceut.4c00685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 09/17/2024]
Abstract
Aqueous solubility is one of the most important physicochemical properties of drug molecules and a major driving force for oral drug absorption. To date, the performance of in silico models for the estimation of solubility for novel chemical space is limited. To investigate possible reasons and remedies for this, the Johnson and Johnson in-house aqueous solubility data with over 40,000 compounds was leveraged. All data were generated through the same high-throughput assay, providing a unique opportunity to explore the relationship between data quality, quantity, and model estimations. Six intrinsic solubility data sets with different sizes and noise levels were generated by making use of three different approaches: (i) inclusion or exclusion of amorphous solid residue, (ii) measured or experimental log D to identify the intrinsic solubility, and (iii) adopting or omitting a quality check process in the data processing workflow. A random forest regressor was trained on the data sets with three different sets of descriptors calculated from RDKit, ADMET predictor, or Mordred, and the performances were evaluated with nested cross-validation as well as ten refined test sets. The models confirm, as expected, that with the same data set size, high-quality data leads to better model performance; however, also, models trained with larger data sets containing analytical variability can give equally accurate estimations compared to models trained with small, clean, and diverse data sets. However, noise introduced by including the presence of amorphous solid postsolubility measurement in the training data set cannot be overcome by increasing data size, as they are introducing a biased systematic positive error in the data set, confirming the importance of critical data review. Finally, two top-performing models were tested on the first test set from the second solubility challenge, achieving RMSE values of 0.74 and 0.72 and log S ± 0.5 of 46 and 48%, respectively. These results demonstrated improved performance compared to those reported in the findings of the competition, highlighting that a single-source curated data set can enhance the prediction of intrinsic solubility.
Collapse
Affiliation(s)
- Jiaxi Zhao
- Department
of Pharmacy, Uppsala University, 751 23 Uppsala, Sweden
| | - Eline Hermans
- Pharmaceutical
& Material Sciences, Janssen Pharmaceutica
NV, B-2340 Beerse, Belgium
| | - Kia Sepassi
- Discovery
Pharmaceutics, Janssen Research & Development,
LLC, La Jolla, California 92121, United States
| | - Christophe Tistaert
- Pharmaceutical
& Material Sciences, Janssen Pharmaceutica
NV, B-2340 Beerse, Belgium
| | | | - Mazen Ahmad
- In
Silico Discovery, Janssen Pharmaceutica
NV, B-2340 Beerse, Belgium
| | - Per Larsson
- Department
of Pharmacy, Uppsala University, 751 23 Uppsala, Sweden
| |
Collapse
|
23
|
Chen ZW, Hua ZL. Effect of Co-exposure to Additional Substances on the Bioconcentration of Per(poly)fluoroalkyl Substances: A Meta-Analysis Based on Hydroponic Experimental Evidence. ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2024; 87:270-286. [PMID: 39367139 DOI: 10.1007/s00244-024-01087-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 08/22/2024] [Indexed: 10/06/2024]
Abstract
A consensus has yet to emerge regarding the bioconcentration responses of per(poly)fluoroalkyl substances under co-exposure with other additional substances in aqueous environments. This study employed a meta-analysis to systematically investigate the aforementioned issues on the basis of 1,085 published datasets of indoor hydroponic simulation experiments. A hierarchical meta-analysis model with an embedded variance covariance matrix was constructed to eliminate the non-independence and shared controls of the data. Overall, the co-exposure resulted in a notable reduction in PFAS bioaccumulation (cumulative effect size, CES = - 0.4287, p < 0.05) and bioconcentration factor (R2 = 0.9507, k < 1, b < 0) in hydroponics. In particular, the inhibition of PFAS bioconcentration induced by dissolved organic matter (percentage form of the effect size, ESP = - 48.98%) was more pronounced than that induced by metal ions (ESP = - 35.54%), particulate matter (ESP = - 24.70%) and persistent organic pollutants (ESP = - 18.66%). A lower AS concentration and a lower concentration ratio of ASs to PFASs significantly promote PFAS bioaccumulation (p < 0.05). The bioaccumulation of PFASs with long chains or high fluoride contents tended to be exacerbated in the presence of ASs. Furthermore, the effect on PFAS bioaccumulation was also significantly dependent on the duration of co-exposure (p < 0.05). The findings of this study provide novel insights into the fate and bioconcentration of PFAS in aquatic environments under co-exposure conditions.
Collapse
Affiliation(s)
- Zi-Wei Chen
- Ministry of Education Key Laboratory of Integrated Regulation and Resource Development on Shallow Lakes, College of Environment, Hohai University, Nanjing, 210098, People's Republic of China
| | - Zu-Lin Hua
- Ministry of Education Key Laboratory of Integrated Regulation and Resource Development on Shallow Lakes, College of Environment, Hohai University, Nanjing, 210098, People's Republic of China.
- Yangtze Institute for Conservation and Development, Nanjing, 210098, People's Republic of China.
| |
Collapse
|
24
|
Mi K, Chou WC, Chen Q, Yuan L, Kamineni VN, Kuchimanchi Y, He C, Monteiro-Riviere NA, Riviere JE, Lin Z. Predicting tissue distribution and tumor delivery of nanoparticles in mice using machine learning models. J Control Release 2024; 374:219-229. [PMID: 39146980 PMCID: PMC11886896 DOI: 10.1016/j.jconrel.2024.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/24/2024] [Accepted: 08/11/2024] [Indexed: 08/17/2024]
Abstract
Nanoparticles (NPs) can be designed for targeted delivery in cancer nanomedicine, but the challenge is a low delivery efficiency (DE) to the tumor site. Understanding the impact of NPs' physicochemical properties on target tissue distribution and tumor DE can help improve the design of nanomedicines. Multiple machine learning and artificial intelligence models, including linear regression, support vector machine, random forest, gradient boosting, and deep neural networks (DNN), were trained and validated to predict tissue distribution and tumor delivery based on NPs' physicochemical properties and tumor therapeutic strategies with the dataset from Nano-Tumor Database. Compared to other machine learning models, the DNN model had superior predictions of DE to tumors and major tissues. The determination coefficients (R2) for the test datasets were 0.41, 0.42, 0.45, 0.79, 0.87, and 0.83 for DE in tumor, heart, liver, spleen, lung, and kidney, respectively. All the R2 and root mean squared error (RMSE) results of the test datasets were similar to the 5-fold cross validation results. Feature importance analysis showed that the core material of NPs played an important role in output predictions among all physicochemical properties. Furthermore, multiple NP formulations with greater DE to the tumor were determined by the DNN model. To facilitate model applications, the final model was converted to a web dashboard. This model could serve as a high-throughput pre-screening tool to support the design of new and efficient nanomedicines with greater tumor DE and serve as an alternative tool to reduce, refine, and partially replace animal experimentation in cancer nanomedicine research.
Collapse
Affiliation(s)
- Kun Mi
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA
| | - Wei-Chun Chou
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA; Department of Environmental Sciences, College of Natural & Agricultural Sciences, University of California, Riverside, CA 92521, USA
| | - Qiran Chen
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA
| | - Long Yuan
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA
| | - Venkata N Kamineni
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA
| | - Yashas Kuchimanchi
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA
| | - Chunla He
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Department of Biostatistics, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA
| | - Nancy A Monteiro-Riviere
- Nanotechnology Innovation Center of Kansas State, Kansas State University, Manhattan, KS 66506, USA; Center for Chemical Toxicology Research and Pharmacokinetics, North Carolina State University, Raleigh, NC 27606, USA
| | - Jim E Riviere
- Center for Chemical Toxicology Research and Pharmacokinetics, North Carolina State University, Raleigh, NC 27606, USA; 1Data Consortium, Kansas State University, Olathe, KS 66061, USA
| | - Zhoumeng Lin
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
25
|
Bharadwaj S, Deepika K, Kumar A, Jaiswal S, Miglani S, Singh D, Fartyal P, Kumar R, Singh S, Singh MP, Gaidhane AM, Kumar B, Jha V. Exploring the Artificial Intelligence and Its Impact in Pharmaceutical Sciences: Insights Toward the Horizons Where Technology Meets Tradition. Chem Biol Drug Des 2024; 104:e14639. [PMID: 39396920 DOI: 10.1111/cbdd.14639] [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: 07/27/2024] [Revised: 09/03/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024]
Abstract
The technological revolutions in computers and the advancement of high-throughput screening technologies have driven the application of artificial intelligence (AI) for faster discovery of drug molecules with more efficiency, and cost-friendly finding of hit or lead molecules. The ability of software and network frameworks to interpret molecular structures' representations and establish relationships/correlations has enabled various research teams to develop numerous AI platforms for identifying new lead molecules or discovering new targets for already established drug molecules. The prediction of biological activity, ADME properties, and toxicity parameters in early stages have reduced the chances of failure and associated costs in later clinical stages, which was observed at a high rate in the tedious, expensive, and laborious drug discovery process. This review focuses on the different AI and machine learning (ML) techniques with their applications mainly focused on the pharmaceutical industry. The applications of AI frameworks in the identification of molecular target, hit identification/hit-to-lead optimization, analyzing drug-receptor interactions, drug repurposing, polypharmacology, synthetic accessibility, clinical trial design, and pharmaceutical developments are discussed in detail. We have also compiled the details of various startups in AI in this field. This review will provide a comprehensive analysis and outline various state-of-the-art AI/ML techniques to the readers with their framework applications. This review also highlights the challenges in this field, which need to be addressed for further success in pharmaceutical applications.
Collapse
Affiliation(s)
- Shruti Bharadwaj
- Center for SeNSE, Indian Institute of Technology Delhi (IIT), New Delhi, India
| | - Kumari Deepika
- Department of Computer Engineering, Pune Institute of Computer Technology, Pune, India
| | - Asim Kumar
- Amity Institute of Pharmacy (AIP), Amity University Haryana, Manesar, India
| | - Shivani Jaiswal
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Shaweta Miglani
- Department of Education, Central University of Punjab, Bathinda, India
| | - Damini Singh
- IES Institute of Pharmacy, IES University, Bhopal, Madhya Pradesh, India
| | - Prachi Fartyal
- Department of Mathematics, Govt PG College Bajpur (US Nagar), Bazpur, Uttarakhand, India
| | - Roshan Kumar
- Department of Microbiology, Graphic Era (Deemed to be University), Dehradun, India
- Department of Microbiology, Central University of Punjab, VPO-Ghudda, Punjab, India
| | - Shareen Singh
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Mahendra Pratap Singh
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Abhay M Gaidhane
- Jawaharlal Nehru Medical College, and Global Health Academy, School of Epidemiology and Public Health, Datta Meghe Institute of Higher Education, Wardha, India
| | - Bhupinder Kumar
- Department of Pharmaceutical Science, Hemvati Nandan Bahuguna Garhwal (A Central) University, Srinagar, Uttarakhand, India
| | - Vibhu Jha
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, Faculty of Life Sciences, University of Bradford, Bradford, UK
| |
Collapse
|
26
|
Zhu Y, Peng J, Xu C, Lan Z. Unsupervised Machine Learning in the Analysis of Nonadiabatic Molecular Dynamics Simulation. J Phys Chem Lett 2024; 15:9601-9619. [PMID: 39270134 DOI: 10.1021/acs.jpclett.4c01751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
The all-atomic full-dimensional-level simulations of nonadiabatic molecular dynamics (NAMD) in large realistic systems has received high research interest in recent years. However, such NAMD simulations normally generate an enormous amount of time-dependent high-dimensional data, leading to a significant challenge in result analyses. Based on unsupervised machine learning (ML) methods, considerable efforts were devoted to developing novel and easy-to-use analysis tools for the identification of photoinduced reaction channels and the comprehensive understanding of complicated molecular motions in NAMD simulations. Here, we tried to survey recent advances in this field, particularly to focus on how to use unsupervised ML methods to analyze the trajectory-based NAMD simulation results. Our purpose is to offer a comprehensive discussion on several essential components of this analysis protocol, including the selection of ML methods, the construction of molecular descriptors, the establishment of analytical frameworks, their advantages and limitations, and persistent challenges.
Collapse
Affiliation(s)
- Yifei Zhu
- MOE Key Laboratory of Environmental Theoretical Chemistry, SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety, School of Environment, South China Normal University, Guangzhou 510006, P. R. China
| | - Jiawei Peng
- MOE Key Laboratory of Environmental Theoretical Chemistry, SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety, School of Environment, South China Normal University, Guangzhou 510006, P. R. China
| | - Chao Xu
- MOE Key Laboratory of Environmental Theoretical Chemistry, SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety, School of Environment, South China Normal University, Guangzhou 510006, P. R. China
| | - Zhenggang Lan
- MOE Key Laboratory of Environmental Theoretical Chemistry, SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety, School of Environment, South China Normal University, Guangzhou 510006, P. R. China
| |
Collapse
|
27
|
Alamoudi JA. Recent advancements toward the incremsent of drug solubility using environmentally-friendly supercritical CO 2: a machine learning perspective. Front Med (Lausanne) 2024; 11:1467289. [PMID: 39286644 PMCID: PMC11402729 DOI: 10.3389/fmed.2024.1467289] [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: 07/19/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Inadequate bioavailability of therapeutic drugs, which is often the consequence of their unacceptable solubility and dissolution rates, is an indisputable operational challenge of pharmaceutical companies due to its detrimental effect on the therapeutic efficacy. Over the recent decades, application of supercritical fluids (SCFs) (mainly SCCO2) has attracted the attentions of many scientists as promising alternative of toxic and environmentally-hazardous organic solvents due to possessing positive advantages like low flammability, availability, high performance, eco-friendliness and safety/simplicity of operation. Nowadays, application of different machine learning (ML) as a versatile, robust and accurate approach for the prediction of different momentous parameters like solubility and bioavailability has been of great attentions due to the non-affordability and time-wasting nature of experimental investigations. The prominent goal of this article is to review the role of different ML-based tools for the prediction of solubility/bioavailability of drugs using SCCO2. Moreover, the importance of solubility factor in the pharmaceutical industry and different possible techniques for increasing the amount of this parameter in poorly-soluble drugs are comprehensively discussed. At the end, the efficiency of SCCO2 for improving the manufacturing process of drug nanocrystals is aimed to be discussed.
Collapse
Affiliation(s)
- Jawaher Abdullah Alamoudi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| |
Collapse
|
28
|
Suriyaamporn P, Pamornpathomkul B, Patrojanasophon P, Ngawhirunpat T, Rojanarata T, Opanasopit P. The Artificial Intelligence-Powered New Era in Pharmaceutical Research and Development: A Review. AAPS PharmSciTech 2024; 25:188. [PMID: 39147952 DOI: 10.1208/s12249-024-02901-y] [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: 04/28/2024] [Accepted: 07/22/2024] [Indexed: 08/17/2024] Open
Abstract
Currently, artificial intelligence (AI), machine learning (ML), and deep learning (DL) are gaining increased interest in many fields, particularly in pharmaceutical research and development, where they assist in decision-making in complex situations. Numerous research studies and advancements have demonstrated how these computational technologies are used in various pharmaceutical research and development aspects, including drug discovery, personalized medicine, drug formulation, optimization, predictions, drug interactions, pharmacokinetics/ pharmacodynamics, quality control/quality assurance, and manufacturing processes. Using advanced modeling techniques, these computational technologies can enhance efficiency and accuracy, handle complex data, and facilitate novel discoveries within minutes. Furthermore, these technologies offer several advantages over conventional statistics. They allow for pattern recognition from complex datasets, and the models, typically developed from data-driven algorithms, can predict a given outcome (model output) from a set of features (model inputs). Additionally, this review discusses emerging trends and provides perspectives on the application of AI with quality by design (QbD) and the future role of AI in this field. Ethical and regulatory considerations associated with integrating AI into pharmaceutical technology were also examined. This review aims to offer insights to researchers, professionals, and others on the current state of AI applications in pharmaceutical research and development and their potential role in the future of research and the era of pharmaceutical Industry 4.0 and 5.0.
Collapse
Affiliation(s)
- Phuvamin Suriyaamporn
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Boonnada Pamornpathomkul
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Prasopchai Patrojanasophon
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Tanasait Ngawhirunpat
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Theerasak Rojanarata
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Praneet Opanasopit
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand.
| |
Collapse
|
29
|
Snow O, Kazemi A, Bhanshali F, Nasiri A, Rozek A, Ester M. Identifying Synergistic Components of Botanical Fungicide Formulations Using Interpretable Graph Neural Networks. J Chem Inf Model 2024; 64:5786-5795. [PMID: 39031079 DOI: 10.1021/acs.jcim.4c00128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2024]
Abstract
Botanical formulations are promising candidates for developing new biopesticides that can protect crops from pests and diseases while reducing harm to the environment. These biopesticides can be combined with permeation enhancer compounds to boost their efficacy against pests and fungal diseases. However, finding synergistic combinations of these compounds is challenging due to the large and complex chemical space. In this paper, we propose a novel deep learning method that can predict the synergy of botanical products and permeation enhancers based on in vitro assay data. Our method uses a weighted combination of component feature vectors to represent the input mixtures, which enables the model to handle a variable number of components and to interpret the contribution of each component to the synergy. We also employ an ensemble of interpretation methods to provide insights into the underlying mechanisms of synergy. We validate our method by testing the predicted synergistic combinations in wet-lab experiments and show that our method can discover novel and effective biopesticides that would otherwise be difficult to find. Our method is generalizable and applicable to other domains, where predicting mixtures of chemical compounds is important.
Collapse
Affiliation(s)
- Oliver Snow
- Terramera, Vancouver, British Columbia V5Y 1K3, Canada
| | - Amirreza Kazemi
- Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | | | - Alyas Nasiri
- Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Annett Rozek
- Terramera, Vancouver, British Columbia V5Y 1K3, Canada
| | - Martin Ester
- Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| |
Collapse
|
30
|
Chen Z, Li N, Zhang P, Li Y, Li X. CardioDPi: An explainable deep-learning model for identifying cardiotoxic chemicals targeting hERG, Cav1.2, and Nav1.5 channels. JOURNAL OF HAZARDOUS MATERIALS 2024; 474:134724. [PMID: 38805819 DOI: 10.1016/j.jhazmat.2024.134724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/08/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024]
Abstract
The cardiotoxic effects of various pollutants have been a growing concern in environmental and material science. These effects encompass arrhythmias, myocardial injury, cardiac insufficiency, and pericardial inflammation. Compounds such as organic solvents and air pollutants disrupt the potassium, sodium, and calcium ion channels cardiac cell membranes, leading to the dysregulation of cardiac function. However, current cardiotoxicity models have disadvantages of incomplete data, ion channels, interpretability issues, and inability of toxic structure visualization. Herein, an interpretable deep-learning model known as CardioDPi was developed, which is capable of discriminating cardiotoxicity induced by the human Ether-à-go-go-related gene (hERG) channel, sodium channel (Na_v1.5), and calcium channel (Ca_v1.5) blockade. External validation yielded promising area under the ROC curve (AUC) values of 0.89, 0.89, and 0.94 for the hERG, Na_v1.5, and Ca_v1.5 channels, respectively. The CardioDPi can be freely accessed on the web server CardioDPipredictor (http://cardiodpi.sapredictor.cn/). Furthermore, the structural characteristics of cardiotoxic compounds were analyzed and structural alerts (SAs) can be extracted using the user-friendly CardioDPi-SAdetector web service (http://cardiosa.sapredictor.cn/). CardioDPi is a valuable tool for identifying cardiotoxic chemicals that are environmental and health risks. Moreover, the SA system provides essential insights for mode-of-action studies concerning cardiotoxic compounds.
Collapse
Affiliation(s)
- Zhaoyang Chen
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China
| | - Na Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China
| | - Pei Zhang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China
| | - Yan Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China
| | - Xiao Li
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250014, China.
| |
Collapse
|
31
|
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
|
32
|
Sheth TS, Acharya F. Optimization and evaluation of modified release solid dosage forms using artificial neural network. Sci Rep 2024; 14:16358. [PMID: 39014107 PMCID: PMC11252257 DOI: 10.1038/s41598-024-67274-5] [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: 01/25/2024] [Accepted: 07/09/2024] [Indexed: 07/18/2024] Open
Abstract
This study aims to optimize and evaluate drug release kinetics of Modified-Release (MR) solid dosage form of Quetiapine Fumarate MR tablets by using the Artificial Neural Networks (ANNs). In training the neural network, the drug contents of Quetiapine Fumarate MR tablet such as Sodium Citrate, Eudragit® L100 55, Eudragit® L30 D55, Lactose Monohydrate, Dicalcium Phosphate (DCP), and Glyceryl Behenate were used as variable input data and Drug Substance Quetiapine Fumarate, Triethyl Citrate, and Magnesium Stearate were used as constant input data for the formulation of the tablet. The in-vitro dissolution profiles of Quetiapine Fumarate MR tablets at ten different time points were used as a target data. Several layers together build the neural network by connecting the input data with the output data via weights, these weights show importance of input nodes. The training process optimises the weights of the drug product excipients to achieve the desired drug release through the simulation process in MATLAB software. The percentage drug release of predicted formulation matched with the manufactured formulation using the similarity factor (f2), which evaluates network efficiency. The ANNs have enormous potential for rapidly optimizing pharmaceutical formulations with desirable performance characteristics.
Collapse
Affiliation(s)
- Tulsi Sagar Sheth
- Department of Applied Sciences and Humanities, Parul Institute of Engineering and Technology, Parul University, Vadodara, Gujarat, 391760, India
- Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, 391760, India
| | - Falguni Acharya
- Department of Applied Sciences and Humanities, Parul Institute of Engineering and Technology, Parul University, Vadodara, Gujarat, 391760, India.
| |
Collapse
|
33
|
Abbas MKG, Rassam A, Karamshahi F, Abunora R, Abouseada M. The Role of AI in Drug Discovery. Chembiochem 2024; 25:e202300816. [PMID: 38735845 DOI: 10.1002/cbic.202300816] [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: 12/03/2023] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/14/2024]
Abstract
The emergence of Artificial Intelligence (AI) in drug discovery marks a pivotal shift in pharmaceutical research, blending sophisticated computational techniques with conventional scientific exploration to break through enduring obstacles. This review paper elucidates the multifaceted applications of AI across various stages of drug development, highlighting significant advancements and methodologies. It delves into AI's instrumental role in drug design, polypharmacology, chemical synthesis, drug repurposing, and the prediction of drug properties such as toxicity, bioactivity, and physicochemical characteristics. Despite AI's promising advancements, the paper also addresses the challenges and limitations encountered in the field, including data quality, generalizability, computational demands, and ethical considerations. By offering a comprehensive overview of AI's role in drug discovery, this paper underscores the technology's potential to significantly enhance drug development, while also acknowledging the hurdles that must be overcome to fully realize its benefits.
Collapse
Affiliation(s)
- M K G Abbas
- Center for Advanced Materials, Qatar University, P.O. Box, 2713, Doha, Qatar
| | - Abrar Rassam
- Secondary Education, Educational Sciences, Qatar University, P.O. Box, 2713, Doha, Qatar
| | - Fatima Karamshahi
- Department of Chemistry and Earth Sciences, Qatar University, P.O. Box, 2713, Doha, Qatar
| | - Rehab Abunora
- Faculty of Medicine, General Medicine and Surgery, Helwan University, Cairo, Egypt
| | - Maha Abouseada
- Department of Chemistry and Earth Sciences, Qatar University, P.O. Box, 2713, Doha, Qatar
| |
Collapse
|
34
|
Branco F, Cunha J, Mendes M, Vitorino C, Sousa JJ. Peptide-Hitchhiking for the Development of Nanosystems in Glioblastoma. ACS NANO 2024; 18:16359-16394. [PMID: 38861272 PMCID: PMC11223498 DOI: 10.1021/acsnano.4c01790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024]
Abstract
Glioblastoma (GBM) remains the epitome of aggressiveness and lethality in the spectrum of brain tumors, primarily due to the blood-brain barrier (BBB) that hinders effective treatment delivery, tumor heterogeneity, and the presence of treatment-resistant stem cells that contribute to tumor recurrence. Nanoparticles (NPs) have been used to overcome these obstacles by attaching targeting ligands to enhance therapeutic efficacy. Among these ligands, peptides stand out due to their ease of synthesis and high selectivity. This article aims to review single and multiligand strategies critically. In addition, it highlights other strategies that integrate the effects of external stimuli, biomimetic approaches, and chemical approaches as nanocatalytic medicine, revealing their significant potential in treating GBM with peptide-functionalized NPs. Alternative routes of parenteral administration, specifically nose-to-brain delivery and local treatment within the resected tumor cavity, are also discussed. Finally, an overview of the significant obstacles and potential strategies to overcome them are discussed to provide a perspective on this promising field of GBM therapy.
Collapse
Affiliation(s)
- Francisco Branco
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Joana Cunha
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Maria Mendes
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - Carla Vitorino
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - João J. Sousa
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
35
|
Pätzmann N, O'Dwyer PJ, Beránek J, Kuentz M, Griffin BT. Predictive computational models for assessing the impact of co-milling on drug dissolution. Eur J Pharm Sci 2024; 198:106780. [PMID: 38697312 DOI: 10.1016/j.ejps.2024.106780] [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: 02/10/2024] [Revised: 04/12/2024] [Accepted: 04/27/2024] [Indexed: 05/04/2024]
Abstract
Co-milling is an effective technique for improving dissolution rate limited absorption characteristics of poorly water-soluble drugs. However, there is a scarcity of models available to forecast the magnitude of dissolution rate improvement caused by co-milling. Therefore, this study endeavoured to quantitatively predict the increase in dissolution by co-milling based on drug properties. Using a biorelevant dissolution setup, a series of 29 structurally diverse and crystalline drugs were screened in co-milled and physically blended mixtures with Polyvinylpyrrolidone K25. Co-Milling Dissolution Ratios after 15 min (COMDR15 min) and 60 min (COMDR60 min) drug release were predicted by variable selection in the framework of a partial least squares (PLS) regression. The model forecasts the COMDR15 min (R2 = 0.82 and Q2 = 0.77) and COMDR60 min (R2 = 0.87 and Q2 = 0.84) with small differences in root mean square errors of training and test sets by selecting four drug properties. Based on three of these selected variables, applicable multiple linear regression equations were developed with a high predictive power of R2 = 0.83 (COMDR15 min) and R2 = 0.84 (COMDR60 min). The most influential predictor variable was the median drug particle size before milling, followed by the calculated drug logD6.5 value, the calculated molecular descriptor Kappa 3 and the apparent solubility of drugs after 24 h dissolution. The study demonstrates the feasibility of forecasting the dissolution rate improvements of poorly water-solube drugs through co-milling. These models can be applied as computational tools to guide formulation in early stage development.
Collapse
Affiliation(s)
- Nicolas Pätzmann
- School of Pharmacy, University College Cork, Cork, Ireland; Department Preformulation and Biopharmacy, Zentiva, k.s., Prague, Czechia
| | | | - Josef Beránek
- Department Preformulation and Biopharmacy, Zentiva, k.s., Prague, Czechia
| | - Martin Kuentz
- Institute of Pharma Technology, University of Applied Sciences and Arts Northwestern Switzerland, Muttenz, Switzerland
| | | |
Collapse
|
36
|
Zhang Y, Watson S, Ramaswamy Y, Singh G. Intravitreal therapeutic nanoparticles for age-related macular degeneration: Design principles, progress and opportunities. Adv Colloid Interface Sci 2024; 329:103200. [PMID: 38788306 DOI: 10.1016/j.cis.2024.103200] [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: 05/11/2024] [Accepted: 05/19/2024] [Indexed: 05/26/2024]
Abstract
Age-related macular degeneration (AMD) is a leading cause of vision loss in the elderly. The current standard treatment for AMD involves frequent intravitreal administrations of therapeutic agents. While effective, this approach presents challenges, including patient discomfort, inconvenience, and the risk of adverse complications. Nanoparticle-based intravitreal drug delivery platforms offer a promising solution to overcome these limitations. These platforms are engineered to target the retina specifically and control drug release, which enhances drug retention, improves drug concentration and bioavailability at the retinal site, and reduces the frequency of injections. This review aims to uncover the design principles guiding the development of highly effective nanoparticle-based intravitreal drug delivery platforms for AMD treatment. By gaining a deeper understanding of the physiology of ocular barriers and the physicochemical properties of nanoparticles, we establish a basis for designing intravitreal nanoparticles to optimize drug delivery and drug retention in the retina. Furthermore, we review recent nanoparticle-based intravitreal therapeutic strategies to highlight their potential in improving AMD treatment efficiency. Lastly, we address the challenges and opportunities in this field, providing insights into the future of nanoparticle-based drug delivery to improve therapeutic outcomes for AMD patients.
Collapse
Affiliation(s)
- Yuhang Zhang
- The School of Biomedical Engineering, Faculty of IT and Engineering, Sydney Nano Institute, The University of Sydney, Camperdown, NSW 2008, Australia
| | - Stephanie Watson
- Faculty of Medicine and Health, Clinical Ophthalmology and Eye Health, Save Sight Institute, The University of Sydney, Camperdown, NSW 2008, Australia
| | - Yogambha Ramaswamy
- The School of Biomedical Engineering, Faculty of IT and Engineering, Sydney Nano Institute, The University of Sydney, Camperdown, NSW 2008, Australia
| | - Gurvinder Singh
- The School of Biomedical Engineering, Faculty of IT and Engineering, Sydney Nano Institute, The University of Sydney, Camperdown, NSW 2008, Australia.
| |
Collapse
|
37
|
Bao Z, Yung F, Hickman RJ, Aspuru-Guzik A, Bannigan P, Allen C. Data-driven development of an oral lipid-based nanoparticle formulation of a hydrophobic drug. Drug Deliv Transl Res 2024; 14:1872-1887. [PMID: 38158474 DOI: 10.1007/s13346-023-01491-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 01/03/2024]
Abstract
Due to its cost-effectiveness, convenience, and high patient adherence, oral drug administration normally remains the preferred approach. Yet, the effective delivery of hydrophobic drugs via the oral route is often hindered by their limited water solubility and first-pass metabolism. To mitigate these challenges, advanced delivery systems such as solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs) have been developed to encapsulate hydrophobic drugs and enhance their bioavailability. However, traditional design methodologies for these complex formulations often present intricate challenges because they are restricted to a relatively narrow design space. Here, we present a data-driven approach for the accelerated design of SLNs/NLCs encapsulating a model hydrophobic drug, cannabidiol, that combines experimental automation and machine learning. A small subset of formulations, comprising 10% of all formulations in the design space, was prepared in-house, leveraging miniaturized experimental automation to improve throughput and decrease the quantity of drug and materials required. Machine learning models were then trained on the data generated from these formulations and used to predict properties of all SLNs/NLCs within this design space (i.e., 1215 formulations). Notably, formulations predicted to be high-performers via this approach were confirmed to significantly enhance the solubility of the drug by up to 3000-fold and prevented degradation of drug. Moreover, the high-performance formulations significantly enhanced the oral bioavailability of the drug compared to both its free form and an over-the-counter version. Furthermore, this bioavailability matched that of a formulation equivalent in composition to the FDA-approved product, Epidiolex®.
Collapse
Affiliation(s)
- Zeqing Bao
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Fion Yung
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Riley J Hickman
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, ON, M5S 1M1, Canada
| | - Alán Aspuru-Guzik
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, M5S 2E4, Canada
- Vector Institute for Artificial Intelligence, Toronto, ON, M5S 1M1, Canada
- Lebovic Fellow, Canadian Institute for Advanced Research (CIFAR), Toronto, ON, M5S 1M1, Canada
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, ON, M5S 3E5, Canada
- Department of Materials Science & Engineering, University of Toronto, Toronto, ON, M5S 3E4, Canada
- CIFAR Artificial Intelligence Research Chair, Vector Institute, Toronto, ON, M5S 1M1, Canada
- Acceleration Consortium, Toronto, ON, M5S 3H6, Canada
| | - Pauric Bannigan
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada.
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada.
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, ON, M5S 3E5, Canada.
- Acceleration Consortium, Toronto, ON, M5S 3H6, Canada.
| |
Collapse
|
38
|
Anchordoquy T, Artzi N, Balyasnikova IV, Barenholz Y, La-Beck NM, Brenner JS, Chan WCW, Decuzzi P, Exner AA, Gabizon A, Godin B, Lai SK, Lammers T, Mitchell MJ, Moghimi SM, Muzykantov VR, Peer D, Nguyen J, Popovtzer R, Ricco M, Serkova NJ, Singh R, Schroeder A, Schwendeman AA, Straehla JP, Teesalu T, Tilden S, Simberg D. Mechanisms and Barriers in Nanomedicine: Progress in the Field and Future Directions. ACS NANO 2024; 18:13983-13999. [PMID: 38767983 PMCID: PMC11214758 DOI: 10.1021/acsnano.4c00182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
In recent years, steady progress has been made in synthesizing and characterizing engineered nanoparticles, resulting in several approved drugs and multiple promising candidates in clinical trials. Regulatory agencies such as the Food and Drug Administration and the European Medicines Agency released important guidance documents facilitating nanoparticle-based drug product development, particularly in the context of liposomes and lipid-based carriers. Even with the progress achieved, it is clear that many barriers must still be overcome to accelerate translation into the clinic. At the recent conference workshop "Mechanisms and Barriers in Nanomedicine" in May 2023 in Colorado, U.S.A., leading experts discussed the formulation, physiological, immunological, regulatory, clinical, and educational barriers. This position paper invites open, unrestricted, nonproprietary discussion among senior faculty, young investigators, and students to trigger ideas and concepts to move the field forward.
Collapse
Affiliation(s)
- Thomas Anchordoquy
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, the University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Natalie Artzi
- Brigham and Woman's Hospital, Department of Medicine, Division of Engineering in Medicine, Harvard Medical School, Boston, Massachusetts 02215, United States
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02215, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02215, United States
| | - Irina V Balyasnikova
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University; Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Yechezkel Barenholz
- Membrane and Liposome Research Lab, IMRIC, Hebrew University Hadassah Medical School, Jerusalem 9112102, Israel
| | - Ninh M La-Beck
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, Texas 79601, United States
| | - Jacob S Brenner
- Departments of Medicine and Pharmacology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Warren C W Chan
- Institute of Biomedical Engineering, University of Toronto, Rosebrugh Building, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Italian Institute of Technology, 16163 Genova, Italy
| | - Agata A Exner
- Departments of Radiology and Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| | - Alberto Gabizon
- The Helmsley Cancer Center, Shaare Zedek Medical Center and The Hebrew University of Jerusalem-Faculty of Medicine, Jerusalem, 9103102, Israel
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Department of Obstetrics and Gynecology, Houston Methodist Hospital, Houston, Texas 77030, United States
- Department of Obstetrics and Gynecology, Weill Cornell Medicine College (WCMC), New York, New York 10065, United States
- Department of Biomedical Engineering, Texas A&M, College Station, Texas 7784,3 United States
| | - Samuel K Lai
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Center for Biohybrid Medical Systems, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - S Moein Moghimi
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, U.K
- Translational and Clinical Research Institute, Faculty of Health and Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, U.K
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Center, Aurora, Colorado 80045, United States
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, The Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Dan Peer
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Juliane Nguyen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rachela Popovtzer
- Faculty of Engineering and the Institute of Nanotechnology & Advanced Materials, Bar-Ilan University, 5290002 Ramat Gan, Israel
| | - Madison Ricco
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, the University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Natalie J Serkova
- Department of Radiology, University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Ravi Singh
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27101, United States
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina 27101, United States
| | - Avi Schroeder
- Department of Chemical Engineering, Technion, Israel Institute of Technology, Haifa 32000, Israel
| | - Anna A Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48108; Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48108, United States
| | - Joelle P Straehla
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts 02115 United States
- Koch Institute for Integrative Cancer Research at MIT, Cambridge Massachusetts 02139 United States
| | - Tambet Teesalu
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Scott Tilden
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, the University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Dmitri Simberg
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, the University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| |
Collapse
|
39
|
Zhou Y, Wang Z, Huang Z, Li W, Chen Y, Yu X, Tang Y, Liu G. In silico prediction of ocular toxicity of compounds using explainable machine learning and deep learning approaches. J Appl Toxicol 2024; 44:892-907. [PMID: 38329145 DOI: 10.1002/jat.4586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/16/2024] [Accepted: 01/16/2024] [Indexed: 02/09/2024]
Abstract
The accurate identification of chemicals with ocular toxicity is of paramount importance in health hazard assessment. In contemporary chemical toxicology, there is a growing emphasis on refining, reducing, and replacing animal testing in safety evaluations. Therefore, the development of robust computational tools is crucial for regulatory applications. The performance of predictive models is heavily reliant on the quality and quantity of data. In this investigation, we amalgamated the most extensive dataset (4901 compounds) sourced from governmental GHS-compliant databases and literature to develop binary classification models of chemical ocular toxicity. We employed 12 molecular representations in conjunction with six machine learning algorithms and two deep learning algorithms to create a series of binary classification models. The findings indicated that the deep learning method GCN outperformed the machine learning models in cross-validation, achieving an impressive AUC of 0.915. However, the top-performing machine learning model (RF-Descriptor) demonstrated excellent performance with an AUC of 0.869 on the test set and was therefore selected as the best model. To enhance model interpretability, we conducted the SHAP method and attention weights analysis. The two approaches offered visual depictions of the relevance of key descriptors and substructures in predicting ocular toxicity of chemicals. Thus, we successfully struck a delicate balance between data quality and model interpretability, rendering our model valuable for predicting and comprehending potential ocular-toxic compounds in the early stages of drug discovery.
Collapse
Affiliation(s)
- Yiqing Zhou
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Ze Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Zejun Huang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Weihua Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yuanting Chen
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xinxin Yu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yun Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Guixia Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
40
|
Li X, Sun Y, Zhou Z, Li J, Liu S, Chen L, Shi Y, Wang M, Zhu Z, Wang G, Lu Q. Deep Learning-Driven Exploration of Pyrroloquinoline Quinone Neuroprotective Activity in Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308970. [PMID: 38454653 PMCID: PMC11095145 DOI: 10.1002/advs.202308970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/15/2024] [Indexed: 03/09/2024]
Abstract
Alzheimer's disease (AD) is a pressing concern in neurodegenerative research. To address the challenges in AD drug development, especially those targeting Aβ, this study uses deep learning and a pharmacological approach to elucidate the potential of pyrroloquinoline quinone (PQQ) as a neuroprotective agent for AD. Using deep learning for a comprehensive molecular dataset, blood-brain barrier (BBB) permeability is predicted and the anti-inflammatory and antioxidative properties of compounds are evaluated. PQQ, identified in the Mediterranean-DASH intervention for a diet that delays neurodegeneration, shows notable BBB permeability and low toxicity. In vivo tests conducted on an Aβ₁₋₄₂-induced AD mouse model verify the effectiveness of PQQ in reducing cognitive deficits. PQQ modulates genes vital for synapse and anti-neuronal death, reduces reactive oxygen species production, and influences the SIRT1 and CREB pathways, suggesting key molecular mechanisms underlying its neuroprotective effects. This study can serve as a basis for future studies on integrating deep learning with pharmacological research and drug discovery.
Collapse
Affiliation(s)
- Xinuo Li
- Jiangsu Provincial Key Laboratory of Drug Metabolism and PharmacokineticsState Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing211166China
| | - Yuan Sun
- Jiangsu Provincial Key Laboratory of Drug Metabolism and PharmacokineticsState Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing211166China
| | - Zheng Zhou
- Department of Computer ScienceRWTH Aachen University52074AachenGermany
| | - Jinran Li
- Jiangsu Provincial Key Laboratory of Drug Metabolism and PharmacokineticsState Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing211166China
| | - Sai Liu
- Jiangsu Provincial Key Laboratory of Drug Metabolism and PharmacokineticsState Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing211166China
| | - Long Chen
- Jiangsu Provincial Key Laboratory of Drug Metabolism and PharmacokineticsState Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing211166China
| | - Yiting Shi
- Jiangsu Provincial Key Laboratory of Drug Metabolism and PharmacokineticsState Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing211166China
| | - Min Wang
- Affiliated Brain Hospital of Nanjing Medical UniversityNanjing210029China
| | - Zheying Zhu
- School of PharmacyThe University of NottinghamNottinghamNG7 2RDUK
| | - Guangji Wang
- Jiangsu Provincial Key Laboratory of Drug Metabolism and PharmacokineticsState Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing211166China
| | - Qiulun Lu
- Jiangsu Provincial Key Laboratory of Drug Metabolism and PharmacokineticsState Key Laboratory of Natural MedicinesChina Pharmaceutical UniversityNanjing211166China
| |
Collapse
|
41
|
Wan K, He J, Shi X. Construction of High Accuracy Machine Learning Interatomic Potential for Surface/Interface of Nanomaterials-A Review. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2305758. [PMID: 37640376 DOI: 10.1002/adma.202305758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/24/2023] [Indexed: 08/31/2023]
Abstract
The inherent discontinuity and unique dimensional attributes of nanomaterial surfaces and interfaces bestow them with various exceptional properties. These properties, however, also introduce difficulties for both experimental and computational studies. The advent of machine learning interatomic potential (MLIP) addresses some of the limitations associated with empirical force fields, presenting a valuable avenue for accurate simulations of these surfaces/interfaces of nanomaterials. Central to this approach is the idea of capturing the relationship between system configuration and potential energy, leveraging the proficiency of machine learning (ML) to precisely approximate high-dimensional functions. This review offers an in-depth examination of MLIP principles and their execution and elaborates on their applications in the realm of nanomaterial surface and interface systems. The prevailing challenges faced by this potent methodology are also discussed.
Collapse
Affiliation(s)
- Kaiwei Wan
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Jianxin He
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Xinghua Shi
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| |
Collapse
|
42
|
Du Y, Song J, Lu L, Yeung E, Givand J, Procopio A, Su Y, Hu G. Design of a Reciprocal Injection Device for Stability Studies of Parenteral Biological Drug Products. J Pharm Sci 2024; 113:1330-1338. [PMID: 38113997 DOI: 10.1016/j.xphs.2023.12.014] [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: 10/01/2023] [Revised: 12/14/2023] [Accepted: 12/14/2023] [Indexed: 12/21/2023]
Abstract
Formulation screening, essential for assessing the impact of physical, chemical, and mechanical stresses on protein stability, plays a critical role in biologics drug product development. This research introduces a Reciprocal Injection Device (RID) designed to accelerate formulation screening by probing protein stability under intensified stress conditions within prefilled syringes. This versatile device is designed to accommodate a broad spectrum of injection parameters and diverse syringe dimensions. A commercial drug product was employed as a model monoclonal antibody formulation. Our findings effectively highlight the efficacy of the RID in assessing concentration-dependent protein stability. This device exhibits significant potential to amplify the influences of interfacial interactions, such as those with buffer salts, excipients, air, metals, and silicone oils, commonly found in combination drug products, and to evaluate the protein stability under varied stresses.
Collapse
Affiliation(s)
- Yong Du
- Analytical Research and Development, Merck & Co., Inc., Rahway, NJ 07065, United States
| | - Jing Song
- Analytical Research and Development, Merck & Co., Inc., Rahway, NJ 07065, United States
| | - Lynn Lu
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, NJ 07065, United States
| | - Edward Yeung
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, NJ 07065, United States
| | - Jeffrey Givand
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, NJ 07065, United States
| | - Adam Procopio
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, NJ 07065, United States
| | - Yongchao Su
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, NJ 07065, United States.
| | - Guangli Hu
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, NJ 07065, United States.
| |
Collapse
|
43
|
Zhou L, Ni C, Liao R, Tang X, Yi T, Ran M, Huang M, Liao R, Zhou X, Qin D, Wang L, Huang F, Xie X, Wan Y, Luo J, Wang Y, Wu J. Activating SRC/MAPK signaling via 5-HT1A receptor contributes to the effect of vilazodone on improving thrombocytopenia. eLife 2024; 13:RP94765. [PMID: 38573820 PMCID: PMC10994662 DOI: 10.7554/elife.94765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024] Open
Abstract
Thrombocytopenia caused by long-term radiotherapy and chemotherapy exists in cancer treatment. Previous research demonstrates that 5-Hydroxtrayptamine (5-HT) and its receptors induce the formation of megakaryocytes (MKs) and platelets. However, the relationships between 5-HT1A receptor (5-HTR1A) and MKs is unclear so far. We screened and investigated the mechanism of vilazodone as a 5-HTR1A partial agonist in promoting MK differentiation and evaluated its therapeutic effect in thrombocytopenia. We employed a drug screening model based on machine learning (ML) to screen the megakaryocytopoiesis activity of Vilazodone (VLZ). The effects of VLZ on megakaryocytopoiesis were verified in HEL and Meg-01 cells. Tg (itga2b: eGFP) zebrafish was performed to analyze the alterations in thrombopoiesis. Moreover, we established a thrombocytopenia mice model to investigate how VLZ administration accelerates platelet recovery and function. We carried out network pharmacology, Western blot, and immunofluorescence to demonstrate the potential targets and pathway of VLZ. VLZ has been predicted to have a potential biological action. Meanwhile, VLZ administration promotes MK differentiation and thrombopoiesis in cells and zebrafish models. Progressive experiments showed that VLZ has a potential therapeutic effect on radiation-induced thrombocytopenia in vivo. The network pharmacology and associated mechanism study indicated that SRC and MAPK signaling are both involved in the processes of megakaryopoiesis facilitated by VLZ. Furthermore, the expression of 5-HTR1A during megakaryocyte differentiation is closely related to the activation of SRC and MAPK. Our findings demonstrated that the expression of 5-HTR1A on MK, VLZ could bind to the 5-HTR1A receptor and further regulate the SRC/MAPK signaling pathway to facilitate megakaryocyte differentiation and platelet production, which provides new insights into the alternative therapeutic options for thrombocytopenia.
Collapse
Affiliation(s)
- Ling Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical UniversityLuZhouChina
| | - Chengyang Ni
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical UniversityLuZhouChina
| | - Ruixue Liao
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical UniversityLuZhouChina
| | - Xiaoqin Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical UniversityLuZhouChina
| | - Taian Yi
- School of Pharmacy, Chengdu University of Traditional Chinese MedicineChengduChina
| | - Mei Ran
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical UniversityLuZhouChina
- School of Basic Medical Sciences, Southwest Medical UniversityLuzhouChina
| | - Miao Huang
- School of Pharmacy, Chengdu University of Traditional Chinese MedicineChengduChina
| | - Rui Liao
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical UniversityLuZhouChina
| | - Xiaogang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical UniversityLuZhouChina
| | - Dalian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical UniversityLuZhouChina
| | - Long Wang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical UniversityLuZhouChina
| | - Feihong Huang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical UniversityLuZhouChina
| | - Xiang Xie
- School of Basic Medical Sciences, Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical UniversityLuzhouChina
| | - Ying Wan
- School of Basic Medical Sciences, Southwest Medical UniversityLuzhouChina
| | - Jiesi Luo
- School of Basic Medical Sciences, Southwest Medical UniversityLuzhouChina
| | - Yiwei Wang
- School of Basic Medical Sciences, Southwest Medical UniversityLuzhouChina
| | - Jianming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical UniversityLuZhouChina
- School of Basic Medical Sciences, Southwest Medical UniversityLuzhouChina
- Education Ministry Key Laboratory of Medical Electrophysiology, Southwest Medical UniversityLuzhouChina
| |
Collapse
|
44
|
Agu PC, Obulose CN. Piquing artificial intelligence towards drug discovery: Tools, techniques, and applications. Drug Dev Res 2024; 85:e22159. [PMID: 38375772 DOI: 10.1002/ddr.22159] [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: 11/08/2023] [Revised: 01/12/2024] [Accepted: 01/29/2024] [Indexed: 02/21/2024]
Abstract
The purpose of this study was to discuss how artificial intelligence (AI) methods have affected the field of drug development. It looks at how AI models and data resources are reshaping the drug development process by offering more affordable and expedient options to conventional approaches. The paper opens with an overview of well-known information sources for drug development. The discussion then moves on to molecular representation techniques that make it possible to convert data into representations that computers can understand. The paper also gives a general overview of the algorithms used in the creation of drug discovery models based on AI. In particular, the paper looks at how AI algorithms might be used to forecast drug toxicity, drug bioactivity, and drug physicochemical properties. De novo drug design, binding affinity prediction, and other AI-based models for drug-target interaction were covered in deeper detail. Modern applications of AI in nanomedicine design and pharmacological synergism/antagonism prediction were also covered. The potential advantages of AI in drug development are highlighted as the evaluation comes to a close. It underlines how AI may greatly speed up and improve the efficiency of drug discovery, resulting in the creation of new and better medicines. To fully realize the promise of AI in drug discovery, the review acknowledges the difficulties that come with its uses in this field and advocates for more study and development.
Collapse
Affiliation(s)
- Peter Chinedu Agu
- Department of Biochemistry, College of Science, Evangel University, Akaeze, Ebonyi State, Nigeria
| | - Chidiebere Nwiboko Obulose
- Department of Computer Sciences, Our Savior Institute of Science, Agriculture, and Technology (OSISATECH Polytechnic), Enugu, Nigeria
| |
Collapse
|
45
|
Hang NT, Long NT, Duy ND, Chien NN, Van Phuong N. Towards safer and efficient formulations: Machine learning approaches to predict drug-excipient compatibility. Int J Pharm 2024; 653:123884. [PMID: 38341049 DOI: 10.1016/j.ijpharm.2024.123884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 01/28/2024] [Accepted: 02/03/2024] [Indexed: 02/12/2024]
Abstract
Predicting drug-excipient compatibility is a critical aspect of pharmaceutical formulation design. In this study, we introduced an innovative approach that leverages machine learning techniques to improve the accuracy of drug-excipient compatibility predictions. Mol2vec and 2D molecular descriptors combined with the stacking technique were used to improve the performance of the model. This approach achieved a significant advancement in the predictive capacity as demonstrated by the accuracy, precision, recall, AUC, and MCC of 0.98, 0.87, 0.88, 0.93 and 0.86, respectively. Using the DE-INTERACT model as the benchmark, our stacking model could remarkably detect drug-excipient incompatibility in 10/12 tested cases, while DE-INTERACT managed to recognize only 3 out of 12 incompatibility cases in the validation experiments. To ensure user accessibility, the trained model was deployed to a user-friendly web platform (URL: https://decompatibility.streamlit.app/). This interactive interface accommodated inputs through various types, including names, PubChem CID, or SMILES strings. It promptly generated compatibility predictions alongside corresponding probability scores. However, the continual refinement of model performance is crucial before applying this model in practice.
Collapse
Affiliation(s)
- Nguyen Thu Hang
- Department of Pharmacognosy, Hanoi University of Pharmacy, Hanoi, Viet Nam
| | - Nguyen Thanh Long
- Department of Pharmacognosy, Hanoi University of Pharmacy, Hanoi, Viet Nam
| | - Nguyen Dang Duy
- Department of Pharmacognosy, Hanoi University of Pharmacy, Hanoi, Viet Nam
| | - Nguyen Ngoc Chien
- National Institute of Pharmaceutical Technology, Hanoi University of Pharmacy, Hanoi, Viet Nam
| | - Nguyen Van Phuong
- Department of Pharmacognosy, Hanoi University of Pharmacy, Hanoi, Viet Nam.
| |
Collapse
|
46
|
Liu Z, Lu T, Qian R, Wang Z, Qi R, Zhang Z. Exploiting Nanotechnology for Drug Delivery: Advancing the Anti-Cancer Effects of Autophagy-Modulating Compounds in Traditional Chinese Medicine. Int J Nanomedicine 2024; 19:2507-2528. [PMID: 38495752 PMCID: PMC10944250 DOI: 10.2147/ijn.s455407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 03/06/2024] [Indexed: 03/19/2024] Open
Abstract
Background Cancer continues to be a prominent issue in the field of medicine, as demonstrated by recent studies emphasizing the significant role of autophagy in the development of cancer. Traditional Chinese Medicine (TCM) provides a variety of anti-tumor agents capable of regulating autophagy. However, the clinical application of autophagy-modulating compounds derived from TCM is impeded by their restricted water solubility and bioavailability. To overcome this challenge, the utilization of nanotechnology has been suggested as a potential solution. Nonetheless, the current body of literature on nanoparticles delivering TCM-derived autophagy-modulating anti-tumor compounds for cancer treatment is limited, lacking comprehensive summaries and detailed descriptions. Methods Up to November 2023, a comprehensive research study was conducted to gather relevant data using a variety of databases, including PubMed, ScienceDirect, Springer Link, Web of Science, and CNKI. The keywords utilized in this investigation included "autophagy", "nanoparticles", "traditional Chinese medicine" and "anticancer". Results This review provides a comprehensive analysis of the potential of nanotechnology in overcoming delivery challenges and enhancing the anti-cancer properties of autophagy-modulating compounds in TCM. The evaluation is based on a synthesis of different classes of autophagy-modulating compounds in TCM, their mechanisms of action in cancer treatment, and their potential benefits as reported in various scholarly sources. The findings indicate that nanotechnology shows potential in enhancing the availability of autophagy-modulating agents in TCM, thereby opening up a plethora of potential therapeutic avenues. Conclusion Nanotechnology has the potential to enhance the anti-tumor efficacy of autophagy-modulating compounds in traditional TCM, through regulation of autophagy.
Collapse
Affiliation(s)
- Zixian Liu
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Tianming Lu
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Ruoning Qian
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Zian Wang
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Ruogu Qi
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| | - Zhengguang Zhang
- School of Medicine, Nanjing University of Chinese Medicine, Jiangsu, Nanjing, People’s Republic of China
| |
Collapse
|
47
|
Duran T, Chaudhuri B. Where Might Artificial Intelligence Be Going in Pharmaceutical Development? Mol Pharm 2024; 21:993-995. [PMID: 38376360 DOI: 10.1021/acs.molpharmaceut.4c00112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
|
48
|
He D, Liu Q, Mi Y, Meng Q, Xu L, Hou C, Wang J, Li N, Liu Y, Chai H, Yang Y, Liu J, Wang L, Hou Y. De Novo Generation and Identification of Novel Compounds with Drug Efficacy Based on Machine Learning. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307245. [PMID: 38204214 PMCID: PMC10962488 DOI: 10.1002/advs.202307245] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/05/2023] [Indexed: 01/12/2024]
Abstract
One of the main challenges in small molecule drug discovery is finding novel chemical compounds with desirable activity. Traditional drug development typically begins with target selection, but the correlation between targets and disease remains to be further investigated, and drugs designed based on targets may not always have the desired drug efficacy. The emergence of machine learning provides a powerful tool to overcome the challenge. Herein, a machine learning-based strategy is developed for de novo generation of novel compounds with drug efficacy termed DTLS (Deep Transfer Learning-based Strategy) by using dataset of disease-direct-related activity as input. DTLS is applied in two kinds of disease: colorectal cancer (CRC) and Alzheimer's disease (AD). In each case, novel compound is discovered and identified in in vitro and in vivo disease models. Their mechanism of actionis further explored. The experimental results reveal that DTLS can not only realize the generation and identification of novel compounds with drug efficacy but also has the advantage of identifying compounds by focusing on protein targets to facilitate the mechanism study. This work highlights the significant impact of machine learning on the design of novel compounds with drug efficacy, which provides a powerful new approach to drug discovery.
Collapse
Affiliation(s)
- Dakuo He
- College of Information Science and EngineeringState Key Laboratory of Synthetical Automation for Process IndustriesNortheastern UniversityShenyang110819China
| | - Qing Liu
- College of Information Science and EngineeringState Key Laboratory of Synthetical Automation for Process IndustriesNortheastern UniversityShenyang110819China
| | - Yan Mi
- Key Laboratory of Bioresource Research and Development of Liaoning ProvinceCollege of Life and Health SciencesNational Frontiers Science Center for Industrial Intelligence and Systems OptimizationNortheastern UniversityShenyang110169China
- Key Laboratory of Data Analytics and Optimization for Smart IndustryMinistry of EducationNortheastern UniversityShenyang110169China
| | - Qingqi Meng
- Key Laboratory of Bioresource Research and Development of Liaoning ProvinceCollege of Life and Health SciencesNational Frontiers Science Center for Industrial Intelligence and Systems OptimizationNortheastern UniversityShenyang110169China
- Key Laboratory of Data Analytics and Optimization for Smart IndustryMinistry of EducationNortheastern UniversityShenyang110169China
| | - Libin Xu
- Key Laboratory of Bioresource Research and Development of Liaoning ProvinceCollege of Life and Health SciencesNational Frontiers Science Center for Industrial Intelligence and Systems OptimizationNortheastern UniversityShenyang110169China
- Key Laboratory of Data Analytics and Optimization for Smart IndustryMinistry of EducationNortheastern UniversityShenyang110169China
| | - Chunyu Hou
- College of Information Science and EngineeringState Key Laboratory of Synthetical Automation for Process IndustriesNortheastern UniversityShenyang110819China
| | - Jinpeng Wang
- College of Information Science and EngineeringState Key Laboratory of Synthetical Automation for Process IndustriesNortheastern UniversityShenyang110819China
| | - Ning Li
- School of Traditional Chinese Materia MedicaKey Laboratory for TCM Material Basis Study and Innovative Drug Development of Shenyang CityShenyang Pharmaceutical UniversityShenyang110016China
| | - Yang Liu
- Key Laboratory of Structure‐Based Drug Design & Discovery of Ministry of EducationShenyang Pharmaceutical UniversityShenyang110016China
| | - Huifang Chai
- School of PharmacyGuizhou University of Traditional Chinese MedicineGuiyang550025China
| | - Yanqiu Yang
- Key Laboratory of Bioresource Research and Development of Liaoning ProvinceCollege of Life and Health SciencesNational Frontiers Science Center for Industrial Intelligence and Systems OptimizationNortheastern UniversityShenyang110169China
- Key Laboratory of Data Analytics and Optimization for Smart IndustryMinistry of EducationNortheastern UniversityShenyang110169China
| | - Jingyu Liu
- Key Laboratory of Bioresource Research and Development of Liaoning ProvinceCollege of Life and Health SciencesNational Frontiers Science Center for Industrial Intelligence and Systems OptimizationNortheastern UniversityShenyang110169China
- Key Laboratory of Data Analytics and Optimization for Smart IndustryMinistry of EducationNortheastern UniversityShenyang110169China
| | - Lihui Wang
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyang110016China
| | - Yue Hou
- Key Laboratory of Bioresource Research and Development of Liaoning ProvinceCollege of Life and Health SciencesNational Frontiers Science Center for Industrial Intelligence and Systems OptimizationNortheastern UniversityShenyang110169China
- Key Laboratory of Data Analytics and Optimization for Smart IndustryMinistry of EducationNortheastern UniversityShenyang110169China
| |
Collapse
|
49
|
Lu A, Williams RO, Maniruzzaman M. 3D printing of biologics-what has been accomplished to date? Drug Discov Today 2024; 29:103823. [PMID: 37949427 DOI: 10.1016/j.drudis.2023.103823] [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: 08/18/2023] [Revised: 10/27/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023]
Abstract
Three-dimensional (3D) printing is a promising approach for the stabilization and delivery of non-living biologics. This versatile tool builds complex structures and customized resolutions, and has significant potential in various industries, especially pharmaceutics and biopharmaceutics. Biologics have become increasingly prevalent in the field of medicine due to their diverse applications and benefits. Stability is the main attribute that must be achieved during the development of biologic formulations. 3D printing could help to stabilize biologics by entrapment, support binding, or crosslinking. Furthermore, gene fragments could be transited into cells during co-printing, when the pores on the membrane are enlarged. This review provides: (i) an introduction to 3D printing technologies and biologics, covering genetic elements, therapeutic proteins, antibodies, and bacteriophages; (ii) an overview of the applications of 3D printing of biologics, including regenerative medicine, gene therapy, and personalized treatments; (iii) information on how 3D printing could help to stabilize and deliver biologics; and (iv) discussion on regulations, challenges, and future directions, including microneedle vaccines, novel 3D printing technologies and artificial-intelligence-facilitated research and product development. Overall, the 3D printing of biologics holds great promise for enhancing human health by providing extended longevity and enhanced quality of life, making it an exciting area in the rapidly evolving field of biomedicine.
Collapse
Affiliation(s)
- Anqi Lu
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Robert O Williams
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Mohammed Maniruzzaman
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38677, USA.
| |
Collapse
|
50
|
Shomope I, Percival KM, Abdel Jabbar NM, Husseini GA. Predicting Calcein Release from Ultrasound-Targeted Liposomes: A Comparative Analysis of Random Forest and Support Vector Machine. Technol Cancer Res Treat 2024; 23:15330338241296725. [PMID: 39539114 PMCID: PMC11561990 DOI: 10.1177/15330338241296725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/29/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE This study presents a comparative analysis of RF and SVM for predicting calcein release from ultrasound-triggered, targeted liposomes under varied low-frequency ultrasound (LFUS) power densities (6.2, 9, and 10 mW/cm2). METHODS Liposomes loaded with calcein and targeted with seven different moieties (cRGD, estrone, folate, Herceptin, hyaluronic acid, lactobionic acid, and transferrin) were synthesized using the thin-film hydration method. The liposomes were characterized using Dynamic Light Scattering and Bicinchoninic Acid assays. Extensive data collection and preprocessing were performed. RF and SVM models were trained and evaluated using mean absolute error (MAE), mean squared error (MSE), coefficient of determination (R²), and the a20 index as performance metrics. RESULTS RF consistently outperformed SVM, achieving R2 scores above 0.96 across all power densities, particularly excelling at higher power densities and indicating a strong correlation with the actual data. CONCLUSION RF outperforms SVM in drug release prediction, though both show strengths and apply based on specific prediction needs.
Collapse
Affiliation(s)
- Ibrahim Shomope
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah 26666, United Arab Emirates
| | - Kelly M. Percival
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah 26666, United Arab Emirates
| | - Nabil M. Abdel Jabbar
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah 26666, United Arab Emirates
| | - Ghaleb A. Husseini
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah PO Box 26666, United Arab Emirates
| |
Collapse
|