1
|
Choi Y, Lee H, Beck BR, Lee B, Lee JH, Kim S, Chun SH, Won HS, Ko YH. Repurposing of the Syk inhibitor fostamatinib using a machine learning algorithm. Exp Ther Med 2025; 29:110. [PMID: 40242601 PMCID: PMC12001310 DOI: 10.3892/etm.2025.12860] [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: 11/07/2024] [Accepted: 02/14/2025] [Indexed: 04/18/2025] Open
Abstract
TAM (TYRO3, AXL, MERTK) receptor tyrosine kinases (RTKs) have intrinsic roles in tumor cell proliferation, migration, chemoresistance, and suppression of antitumor immunity. The overexpression of TAM RTKs is associated with poor prognosis in various types of cancer. Single-target agents of TAM RTKs have limited efficacy because of an adaptive feedback mechanism resulting from the cooperation of TAM family members. This suggests that multiple targeting of members has the potential for a more potent anticancer effect. The present study used a deep-learning based drug-target interaction (DTI) prediction model called molecule transformer-DTI (MT-DTI) to identify commercially available drugs that may inhibit the three members of TAM RTKs. The results showed that fostamatinib, a spleen tyrosine kinase (Syk) inhibitor, could inhibit the three receptor kinases of the TAM family with an IC50 <1 µM. Notably, no other Syk inhibitors were predicted by the MT-DTI model. To verify this result, this study performed in vitro studies with various types of cancer cell lines. Consistent with the DTI results, this study observed that fostamatinib suppressed cell proliferation by inhibiting TAM RTKs, while other Syk inhibitors showed no inhibitory activity. These results suggest that fostamatinib could exhibit anticancer activity as a pan-TAM inhibitor. Taken together, these findings demonstrated that this artificial intelligence model could be effectively used for drug repurposing and repositioning. Furthermore, by identifying its novel mechanism of action, this study confirmed the potential for fostamatinib to expand its indications as a TAM inhibitor.
Collapse
Affiliation(s)
| | - Heejin Lee
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Bo Ram Beck
- Deargen Inc., Daejeon 35220, Republic of Korea
| | - Bora Lee
- Deargen Inc., Daejeon 35220, Republic of Korea
| | - Ji Hyun Lee
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Oncology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Seoree Kim
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Oncology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sang Hoon Chun
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Oncology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hye Sung Won
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Oncology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Yoon Ho Ko
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Oncology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
2
|
Wang L, Zhang T, Yang X, Mo Q, Ran M, Li R, Yang B, Shen H, Li Q, Li Z, Jiang N, Zeng J, Xie X, He S, Huang F, Zhang C, Luo J, Wu J. Multimodal discovery of bavachinin A: A natural FLT3 agonist for treating thrombocytopenia. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156597. [PMID: 40058315 DOI: 10.1016/j.phymed.2025.156597] [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: 04/04/2024] [Revised: 11/25/2024] [Accepted: 02/28/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Radiation-induced thrombocytopenia (RIT) poses a serious risk to patients with cancer undergoing radiotherapy and leads to hemorrhage and mortality. Unfortunately, effective treatment options for RIT are currently limited. PURPOSE This study aimed to discover active compound from Fructus Psoraleae, a traditional Chinese medicine recognized for its hemostatic properties, and to elucidate its mechanism of action in the treatment of RIT. METHODS The efficacy of Fructus Psoraleae in treating thrombocytopenia was assessed using network pharmacology. A drug-screening model was built using a naive Bayes algorithm to determine the effective compounds in Fructus Psoraleae. Giemsa staining and flow cytometry were used to evaluate the effects of bavachinin A on megakaryocytes (MK) differentiation. RIT and thrombopoietin (TPO) receptor (c-MPL) knockout (c-MPL-/-) mice were used to assess the therapeutic efficacy of bavachinin A in mitigating thrombocytopenia. Tg (cd41:eGFP) zebrafish were used to investigate the effect of bavachinin A on thrombopoiesis. RNA sequencing (RNA-seq), molecular docking simulations, molecular dynamics simulations, drug affinity responsive target stability assay (DARTS), and biolayer interferometry (BLI) were used to elucidate the molecular mechanisms of action of bavachinin A against thrombocytopenia. RESULTS In silico analysis using a drug screening model identified bavachinin A as promising candidate compound derived from Fructus Psoraleae. In vitro experiments demonstrated that Bavachinin A induced MK differentiation. In vivo experiments revealed that bavachinin A augmented platelet levels and improved coagulation in RIT mice, facilitated megakaryopoiesis and platelet levels in c-MPL-/- mice, and accelerated thrombopoiesis in zebrafish. Furthermore, RNA-seq, molecular docking simulations, molecular dynamics simulations, DARTS, and BLI demonstrated that bavachinin A bound directly to fms-like tyrosine kinase 3 (FLT3). Notably, blocking FLT3 or phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway hindered bavachinin-A-induced MK differentiation. However, repressing the TPO/c-MPL signaling pathway had no significant effect. CONCLUSION Bavachinin A promotes MK differentiation and thrombopoiesis by directly binding to FLT3 and activating PI3K/Akt signaling. Importantly, this effect was not dependent on the conventional TPO/c-MPL signaling pathway. This study underscores the translational potential of bavachinin A as a promising novel therapeutic for thrombocytopenia, offering novel insights into TPO-independent mechanisms of thrombopoiesis and establishing a robust multimodal approach for drug discovery.
Collapse
Affiliation(s)
- Long Wang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Ting Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xin Yang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Qi Mo
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Mei Ran
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Rong Li
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan, 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, The School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Bo Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Hongping Shen
- Clinical Trial Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Qinyao Li
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Zhichao Li
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Nan Jiang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jing Zeng
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xiang Xie
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Siyu He
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Feihong Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Chunxiang Zhang
- Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Jiesi Luo
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Jianming Wu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China; School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China; Education Ministry Key Laboratory of Medical Electrophysiology, Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
3
|
Siddiqui NF, Vishwakarma P, Thakur S, Jadhav HR. Bioactivity predictions and virtual screening using machine learning predictive model. J Biomol Struct Dyn 2025; 43:3909-3928. [PMID: 38217308 DOI: 10.1080/07391102.2023.2300132] [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: 09/20/2023] [Accepted: 12/23/2023] [Indexed: 01/15/2024]
Abstract
Recently, there has been significant attention on machine learning algorithms for predictive modeling. Prediction models for enzyme inhibitors are limited, and it is essential to account for chemical biases while developing them. The lack of repeatability in available models and chemical bias issues constrain drug discovery and development. A new prediction model for enzyme inhibitors has been developed, and the model efficacy was checked using Dipeptidyl peptidase 4 (DPP-4) inhibitors. A Python script was prepared and can be provided for personal use upon request. Among various machine learning algorithms, it was found that Random Forest offers the best accuracy. Two models were compared, one with diverse training and test data and the other with a random split. It was concluded that machine learning predictive models based on the Murcko scaffold can address chemical bias concerns. In-silico screening of the Drug Bank database identified two molecules against DPP-4, which are previously proven hit molecules. The approach was further validated through molecular docking studies and molecular dynamics simulations, demonstrating the credibility and relevance of the developed model for future investigations and potential translation into clinical applications.
Collapse
Affiliation(s)
- Noor Fatima Siddiqui
- Department of Pharmacy, Pharmaceutical Chemistry Research Laboratory, Birla Institute of Technology and Science Pilani, Pilani, RJ, India
| | - Pinky Vishwakarma
- Department of Pharmacy, Pharmaceutical Chemistry Research Laboratory, Birla Institute of Technology and Science Pilani, Pilani, RJ, India
| | - Shikha Thakur
- Department of Pharmacy, Pharmaceutical Chemistry Research Laboratory, Birla Institute of Technology and Science Pilani, Pilani, RJ, India
| | - Hemant R Jadhav
- Department of Pharmacy, Pharmaceutical Chemistry Research Laboratory, Birla Institute of Technology and Science Pilani, Pilani, RJ, India
| |
Collapse
|
4
|
Wang L, He X, Shen Y, Chen J, Chen Y, Zhou Z, Xu X. A Novel Ferroptosis-Related Gene Prognosis Signature and Identifying Atorvastatin as a Potential Therapeutic Agent for Hepatocellular Carcinoma. Curr Issues Mol Biol 2025; 47:201. [PMID: 40136455 PMCID: PMC11940908 DOI: 10.3390/cimb47030201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/09/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Among the most common malignant tumors, hepatocellular carcinoma (HCC) is a primary liver cancer type that has a high mortality rate. HCC often presents insidiously, is prone to recurrence, and has limited treatment efficacy. Ferroptosis regulates tumorigenesis, progression, and metastasis, which is a novel form of iron-dependent cell death. Numerous studies suggest that HCC is sensitive to ferroptosis, indicating that targeted therapies aimed at inducing ferroptosis may represent a promising new approach to cancer treatment. This study aims to find genes associated with HCC and ferroptosis, as well as to screen for potential agents that may cause ferroptosis in HCC. Transcriptome and clinical sample data were obtained from the TCGA database to identify differentially expressed genes related to ferroptosis. Using various regression and survival analysis techniques, we developed a prognostic model based on four core genes and evaluated its predictive potential. Subsequently, we screened for potential therapeutic agents in the Connective Map (CMap) database, designated as compound Atorvastatin, based on differential genes from two risk groups and related to ferroptosis. Through experiments conducted in vivo and in vitro, we demonstrated that Atorvastatin can induce ferroptosis in HCC cells while inhibiting their growth and migration. In conclusion, this research targets ferroptosis therapy and provides new insights for improving the prediction and prevention of HCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ximing Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China; (L.W.); (X.H.); (Y.S.); (J.C.); (Y.C.); (Z.Z.)
| |
Collapse
|
5
|
Chaudhary S, Rahate K, Mishra S. Harnessing machine learning for rational drug design. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2025; 103:209-230. [PMID: 40175042 DOI: 10.1016/bs.apha.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
A crucial part of biomedical research is drug discovery, which aims to find and create innovative medical treatments for a range of illnesses. However, there are intrinsic obstacles to the traditional approach of discovering novel medications, including high prices, lengthy turnaround times, and poor clinical trial success rates. In recent times, the use of designing algorithms for machine learning has become a groundbreaking way to improve and optimise many stages of medication development. An outline of the quickly developing area of machine learning algorithms for drug discovery is given in this review, emphasising how revolutionary treatment development might be. To effectively get a novel medication into the market, modern medicinal development often involves many interconnected stages. The use of computational tools has become more and more crucial in reducing the time and cost involved in the investigation and creation of new medications. Our latest efforts to combine molecular modelling as well as machine learning to create the computational resources for designing modulators utilising a sensible design influenced by the pocket process that targets protein-protein interactions via AlphaSpace are reviewed in this Perspective. A significant shift in pharmaceutical research has occurred with the introduction of AI in drug discovery, which combines cutting-edge computer techniques with conventional scientific investigation to address enduring problems. By highlighting significant advancements and methodologies, this review paper elucidates the many applications of AI throughout several stages of drug discovery.
Collapse
Affiliation(s)
- Sandhya Chaudhary
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Kalpana Rahate
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India.
| | - Shuchita Mishra
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
6
|
Yuan Y, Li Y, Li G, Lei L, Huang X, Li M, Yao Y. Intelligent Design of Lipid Nanoparticles for Enhanced Gene Therapeutics. Mol Pharm 2025; 22:1142-1159. [PMID: 39878334 DOI: 10.1021/acs.molpharmaceut.4c00925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Lipid nanoparticles (LNPs) are an effective delivery system for gene therapeutics. By optimizing their formulation, the physiochemical properties of LNPs can be tailored to improve tissue penetration, cellular uptake, and precise targeting. The application of these targeted delivery strategies within the LNP framework ensures efficient delivery of therapeutic agents to specific organs or cell types, thereby maximizing therapeutic efficacy. In the realm of genome editing, LNPs have emerged as a potent vehicle for delivering CRISPR/Cas components, offering significant advantages such as high in vivo efficacy. The incorporation of machine learning into the optimization of LNP platforms for gene therapeutics represents a significant advancement, harnessing its predictive capabilities to substantially accelerate the research and development process. This review highlights the dynamic evolution of LNP technology, which is expected to drive transformative progress in the field of gene therapy.
Collapse
Affiliation(s)
- Yichen Yuan
- ZJU-Hangzhou Global Scientific and Technological Innovation Canter, Zhejiang University, Hangzhou, Zhejiang 311215, China
- Research Center for Life Sciences Computing, Zhejiang Lab, Hangzhou, Zhejiang 311121, China
| | - Ying Li
- Research Center for Space Computing System, Zhejiang Lab, Hangzhou, Zhejiang 311121, China
| | - Guo Li
- ZJU-Hangzhou Global Scientific and Technological Innovation Canter, Zhejiang University, Hangzhou, Zhejiang 311215, China
| | - Liqun Lei
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 311100, China
| | - Xingxu Huang
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 311100, China
| | - Ming Li
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai 201102, China
| | - Yuan Yao
- ZJU-Hangzhou Global Scientific and Technological Innovation Canter, Zhejiang University, Hangzhou, Zhejiang 311215, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
- Zhejiang Key Laboratory of Intelligent Manufacturing for Functional Chemicals, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou 311215, China
| |
Collapse
|
7
|
Chen B, Liu S, Xia H, Li X, Zhang Y. Computer-Aided Drug Design in Research on Chinese Materia Medica: Methods, Applications, Advantages, and Challenges. Pharmaceutics 2025; 17:315. [PMID: 40142979 PMCID: PMC11945071 DOI: 10.3390/pharmaceutics17030315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Chinese materia medica (CMM) refers to the medicinal substances used in traditional Chinese medicine. In recent years, CMM has become globally prevalent, and scientific research on CMM has increasingly garnered attention. Computer-aided drug design (CADD) has been employed in Western medicine research for many years, contributing significantly to its progress. However, the role of CADD in CMM research has not been systematically reviewed. This review briefly introduces CADD methods in CMM research from the perspectives of computational chemistry (including quantum chemistry, molecular mechanics, and quantum mechanics/molecular mechanics) and informatics (including cheminformatics, bioinformatics, and data mining). Then, it provides an exhaustive discussion of the applications of these CADD methods in CMM research through rich cases. Finally, the review outlines the advantages and challenges of CADD in CMM research. In conclusion, despite the current challenges, CADD still offers unique advantages over traditional experiments. With the development of the CMM industry and computer science, especially driven by artificial intelligence, CADD is poised to play an increasingly pivotal role in advancing CMM research.
Collapse
Affiliation(s)
- Ban Chen
- Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Centre of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan 430068, China; (B.C.); (S.L.); (H.X.)
| | - Shuangshuang Liu
- Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Centre of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan 430068, China; (B.C.); (S.L.); (H.X.)
| | - Huiyin Xia
- Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Centre of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan 430068, China; (B.C.); (S.L.); (H.X.)
| | - Xican Li
- School of Chinese Herbal Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China;
| | - Yingqing Zhang
- Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Centre of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan 430068, China; (B.C.); (S.L.); (H.X.)
| |
Collapse
|
8
|
You K, Binte Mohamed Yazid N, Chong LM, Hooi L, Wang P, Zhuang I, Chua S, Lim E, Kok AZX, Marimuthu K, Vasoo S, Ng OT, Chan CEZ, Chow EKH, Ho D. Flash optimization of drug combinations for Acinetobacter baumannii with IDentif.AI-AMR. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:12. [PMID: 39984645 PMCID: PMC11845484 DOI: 10.1038/s44259-025-00079-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 01/15/2025] [Indexed: 02/23/2025]
Abstract
Antimicrobial resistance (AMR) is an emerging threat to global public health. Specifically, Acinetobacter baumannii (A. baumannii), one of the main pathogens driving the rise of nosocomial infections, is a Gram-negative bacillus that displays intrinsic resistance mechanisms and can also develop resistance by acquiring AMR genes from other bacteria. More importantly, it is resistant to nearly 90% of standard of care (SOC) antimicrobial treatments, resulting in unsatisfactory clinical outcomes and a high infection-associated mortality rate of over 30%. Currently, there is a growing challenge to sustainably develop novel antimicrobials in this ever-expanding arms race against AMR. Therefore, a sustainable workflow that properly manages healthcare resources to ultra-rapidly design optimal drug combinations for effective treatment is needed. In this study, the IDentif.AI-AMR platform was harnessed to pinpoint effective regimens against four A. baumannii clinical isolates from a pool of nine US FDA-approved drugs. Notably, IDentif.AI-pinpointed ampicillin-sulbactam/cefiderocol and cefiderocol/polymyxin B/rifampicin combinations were able to achieve 93.89 ± 5.95% and 92.23 ± 11.89% inhibition against the bacteria, respectively, and they may diversify the reservoir of treatment options for the indication. In addition, polymyxin B in combination with rifampicin exhibited broadly applicable efficacy and strong synergy across all tested clinical isolates, representing a potential treatment strategy for A. baumannii. IDentif.AI-pinpointed combinations may potentially serve as alternative treatment strategies for A. baumannii.
Collapse
Affiliation(s)
- Kui You
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
- Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore
| | | | - Li Ming Chong
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
- Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore
| | - Lissa Hooi
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Peter Wang
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
- Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore
| | - Isaiah Zhuang
- Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Stephen Chua
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Ethan Lim
- Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore
| | - Alrick Zi Xin Kok
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | | | - Shawn Vasoo
- National Centre for Infectious Diseases (NCID), Singapore, Singapore.
| | - Oon Tek Ng
- National Centre for Infectious Diseases (NCID), Singapore, Singapore.
| | - Conrad E Z Chan
- National Centre for Infectious Diseases (NCID), Singapore, Singapore.
| | - Edward Kai-Hua Chow
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore.
- Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Dean Ho
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore.
- Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- The Bia-Echo Asia Centre for Reproductive Longevity and Equality (ACRLE), National University of Singapore, Singapore, Singapore.
| |
Collapse
|
9
|
Harkos C, Hadjigeorgiou AG, Voutouri C, Kumar AS, Stylianopoulos T, Jain RK. Using mathematical modelling and AI to improve delivery and efficacy of therapies in cancer. Nat Rev Cancer 2025:10.1038/s41568-025-00796-w. [PMID: 39972158 DOI: 10.1038/s41568-025-00796-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/30/2025] [Indexed: 02/21/2025]
Abstract
Mathematical modelling has proven to be a valuable tool in predicting the delivery and efficacy of molecular, antibody-based, nano and cellular therapy in solid tumours. Mathematical models based on our understanding of the biological processes at subcellular, cellular and tissue level are known as mechanistic models that, in turn, are divided into continuous and discrete models. Continuous models are further divided into lumped parameter models - for describing the temporal distribution of medicine in tumours and normal organs - and distributed parameter models - for studying the spatiotemporal distribution of therapy in tumours. Discrete models capture interactions at the cellular and subcellular levels. Collectively, these models are useful for optimizing the delivery and efficacy of molecular, nanoscale and cellular therapy in tumours by incorporating the biological characteristics of tumours, the physicochemical properties of drugs, the interactions among drugs, cancer cells and various components of the tumour microenvironment, and for enabling patient-specific predictions when combined with medical imaging. Artificial intelligence-based methods, such as machine learning, have ushered in a new era in oncology. These data-driven approaches complement mechanistic models and have immense potential for improving cancer detection, treatment and drug discovery. Here we review these diverse approaches and suggest ways to combine mechanistic and artificial intelligence-based models to further improve patient treatment outcomes.
Collapse
Affiliation(s)
- Constantinos Harkos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Andreas G Hadjigeorgiou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Ashwin S Kumar
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Ambreen S, Umar M, Noor A, Jain H, Ali R. Advanced AI and ML frameworks for transforming drug discovery and optimization: With innovative insights in polypharmacology, drug repurposing, combination therapy and nanomedicine. Eur J Med Chem 2025; 284:117164. [PMID: 39721292 DOI: 10.1016/j.ejmech.2024.117164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/24/2024] [Accepted: 11/27/2024] [Indexed: 12/28/2024]
Abstract
Artificial Intelligence (AI) and Machine Learning (ML) are transforming drug discovery by overcoming traditional challenges like high costs, time-consuming, and frequent failures. AI-driven approaches streamline key phases, including target identification, lead optimization, de novo drug design, and drug repurposing. Frameworks such as deep neural networks (DNNs), convolutional neural networks (CNNs), and deep reinforcement learning (DRL) models have shown promise in identifying drug targets, optimizing delivery systems, and accelerating drug repurposing. Generative adversarial networks (GANs) and variational autoencoders (VAEs) aid de novo drug design by creating novel drug-like compounds with desired properties. Case studies, such as DDR1 kinase inhibitors designed using generative models and CDK20 inhibitors developed via structure-based methods, highlight AI's ability to produce highly specific therapeutics. Models like SNF-CVAE and DeepDR further advance drug repurposing by uncovering new therapeutic applications for existing drugs. Advanced ML algorithms enhance precision in predicting drug efficacy, toxicity, and ADME-Tox properties, reducing development costs and improving drug-target interactions. AI also supports polypharmacology by optimizing multi-target drug interactions and enhances combination therapy through predictions of drug synergies and antagonisms. In nanomedicine, AI models like CURATE.AI and the Hartung algorithm optimize personalized treatments by predicting toxicological risks and real-time dosing adjustments with high accuracy. Despite its potential, challenges like data quality, model interpretability, and ethical concerns must be addressed. High-quality datasets, transparent models, and unbiased algorithms are essential for reliable AI applications. As AI continues to evolve, it is poised to revolutionize drug discovery and personalized medicine, advancing therapeutic development and patient care.
Collapse
Affiliation(s)
- Subiya Ambreen
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Mohammad Umar
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Aaisha Noor
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Himangini Jain
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India
| | - Ruhi Ali
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSRU, Pushp Vihar, New Delhi, 110017, India.
| |
Collapse
|
11
|
Bai X, Zhang X. Artificial Intelligence-Powered Materials Science. NANO-MICRO LETTERS 2025; 17:135. [PMID: 39912967 PMCID: PMC11803041 DOI: 10.1007/s40820-024-01634-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 12/11/2024] [Indexed: 02/07/2025]
Abstract
The advancement of materials has played a pivotal role in the advancement of human civilization, and the emergence of artificial intelligence (AI)-empowered materials science heralds a new era with substantial potential to tackle the escalating challenges related to energy, environment, and biomedical concerns in a sustainable manner. The exploration and development of sustainable materials are poised to assume a critical role in attaining technologically advanced solutions that are environmentally friendly, energy-efficient, and conducive to human well-being. This review provides a comprehensive overview of the current scholarly progress in artificial intelligence-powered materials science and its cutting-edge applications. We anticipate that AI technology will be extensively utilized in material research and development, thereby expediting the growth and implementation of novel materials. AI will serve as a catalyst for materials innovation, and in turn, advancements in materials innovation will further enhance the capabilities of AI and AI-powered materials science. Through the synergistic collaboration between AI and materials science, we stand to realize a future propelled by advanced AI-powered materials.
Collapse
Affiliation(s)
- Xiaopeng Bai
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, 999077, People's Republic of China
- Department of Physics, The Chinese University of Hong Kong, Shatin, Hong Kong, 999077, People's Republic of China
| | - Xingcai Zhang
- World Tea Organization, Cambridge, MA, 02139, USA.
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
12
|
Mswahili ME, Hwang J, Rajapakse JC, Jo K, Jeong YS. Positional embeddings and zero-shot learning using BERT for molecular-property prediction. J Cheminform 2025; 17:17. [PMID: 39910649 PMCID: PMC11800558 DOI: 10.1186/s13321-025-00959-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/18/2025] [Indexed: 02/07/2025] Open
Abstract
Recently, advancements in cheminformatics such as representation learning for chemical structures, deep learning (DL) for property prediction, data-driven discovery, and optimization of chemical data handling, have led to increased demands for handling chemical simplified molecular input line entry system (SMILES) data, particularly in text analysis tasks. These advancements have driven the need to optimize components like positional encoding and positional embeddings (PEs) in transformer model to better capture the sequential and contextual information embedded in molecular representations. SMILES data represent complex relationships among atoms or elements, rendering them critical for various learning tasks within the field of cheminformatics. This study addresses the critical challenge of encoding complex relationships among atoms in SMILES strings to explore various PEs within the transformer-based framework to increase the accuracy and generalization of molecular property predictions. The success of transformer-based models, such as the bidirectional encoder representations from transformer (BERT) models, in natural language processing tasks has sparked growing interest from the domain of cheminformatics. However, the performance of these models during pretraining and fine-tuning is significantly influenced by positional information such as PEs, which help in understanding the intricate relationships within sequences. Integrating position information within transformer architectures has emerged as a promising approach. This encoding mechanism provides essential supervision for modeling dependencies among elements situated at different positions within a given sequence. In this study, we first conduct pretraining experiments using various PEs to explore diverse methodologies for incorporating positional information into the BERT model for chemical text analysis using SMILES strings. Next, for each PE, we fine-tune the best-performing BERT (masked language modeling) model on downstream tasks for molecular-property prediction. Here, we use two molecular representations, SMILES and DeepSMILES, to comprehensively assess the potential and limitations of the PEs in zero-shot learning analysis, demonstrating the model's proficiency in predicting properties of unseen molecular representations in the context of newly proposed and existing datasets.Scientific contributionThis study explores the unexplored potential of PEs using BERT model for molecular property prediction. The study involved pretraining and fine-tuning the BERT model on various datasets related to COVID-19, bioassay data, and other molecular and biological properties using SMILES and DeepSMILES representations. The study details the pretraining architecture, fine-tuning datasets, and the performance of the BERT model with different PEs. It also explores zero-shot learning analysis and the model's performance on various classification and regression tasks. In this study, newly proposed datasets from different domains were introduced during fine-tuning in addition to the existing and commonly used datasets. The study highlights the robustness of the BERT model in predicting chemical properties and its potential applications in cheminformatics and bioinformatics.
Collapse
Affiliation(s)
- Medard Edmund Mswahili
- Department of Computer Engineering, Chungbuk National University, Cheongju, 28644, South Korea
| | - JunHa Hwang
- Department of Computer Engineering, Chungbuk National University, Cheongju, 28644, South Korea
| | - Jagath C Rajapakse
- School of Computer Science and Engineering, Nanyang Technological University, Singapore, Singapore
| | - Kyuri Jo
- Department of Computer Engineering, Chungbuk National University, Cheongju, 28644, South Korea.
| | - Young-Seob Jeong
- Department of Computer Engineering, Chungbuk National University, Cheongju, 28644, South Korea.
| |
Collapse
|
13
|
Li VOK, Han Y, Kaistha T, Zhang Q, Downey J, Gozes I, Lam JCK. DeepDrug as an expert guided and AI driven drug repurposing methodology for selecting the lead combination of drugs for Alzheimer's disease. Sci Rep 2025; 15:2093. [PMID: 39814937 PMCID: PMC11735786 DOI: 10.1038/s41598-025-85947-7] [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: 09/25/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025] Open
Abstract
Alzheimer's Disease (AD) significantly aggravates human dignity and quality of life. While newly approved amyloid immunotherapy has been reported, effective AD drugs remain to be identified. Here, we propose a novel AI-driven drug-repurposing method, DeepDrug, to identify a lead combination of approved drugs to treat AD patients. DeepDrug advances drug-repurposing methodology in four aspects. Firstly, it incorporates expert knowledge to extend candidate targets to include long genes, immunological and aging pathways, and somatic mutation markers that are associated with AD. Secondly, it incorporates a signed directed heterogeneous biomedical graph encompassing a rich set of nodes and edges, and node/edge weighting to capture crucial pathways associated with AD. Thirdly, it encodes the weighted biomedical graph through a Graph Neural Network into a new embedding space to capture the granular relationships across different nodes. Fourthly, it systematically selects the high-order drug combinations via diminishing return-based thresholds. A five-drug lead combination, consisting of Tofacitinib, Niraparib, Baricitinib, Empagliflozin, and Doxercalciferol, has been selected from the top drug candidates based on DeepDrug scores to achieve the maximum synergistic effect. These five drugs target neuroinflammation, mitochondrial dysfunction, and glucose metabolism, which are all related to AD pathology. DeepDrug offers a novel AI-and-big-data, expert-guided mechanism for new drug combination discovery and drug-repurposing across AD and other neuro-degenerative diseases, with immediate clinical applications.
Collapse
Affiliation(s)
- Victor O K Li
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China.
| | - Yang Han
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| | - Tushar Kaistha
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| | - Qi Zhang
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| | - Jocelyn Downey
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| | - Illana Gozes
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Jacqueline C K Lam
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
14
|
Qin Y, Cui F, Lu Y, Yang P, Gou W, Tang Z, Lu S, Zhou HS, Luo G, Lyu X, Zhang Q. Toward precision medicine: End-to-end design and construction of integrated microneedle-based theranostic systems. J Control Release 2025; 377:354-375. [PMID: 39577466 DOI: 10.1016/j.jconrel.2024.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 11/09/2024] [Indexed: 11/24/2024]
Abstract
With the growing demand for precision medicine and advancements in microneedle technology, microneedle-based drug delivery systems have evolved into integrated theranostic platforms. However, the development of these systems is currently limited by the absence of clear conclusions and standardized construction strategies. The end-to-end concept offers an innovative approach to theranostic systems by creating a seamless process that integrates target sampling, sensing, analysis, and on-demand drug delivery. This approach optimizes each step based on data from the others, effectively eliminating the traditional separation between drug delivery and disease monitoring. Furthermore, by incorporating artificial intelligence and machine learning, these systems can enhance reliability and efficiency in disease management, paving the way for more personalized and effective healthcare solutions. Based on the concept of end-to-end and recent advancements in theranostic systems, nanomaterials, electronic components, micro-composites, and data science, we propose a modular strategy for constructing integrated microneedle-based theranostic systems by detailing the methods and functions of each critical component, including monitoring, decision-making, and on-demand drug delivery units, though the total number of units might vary depending on the specific application. Notably, decision-making units are emerging trends for fully automatic and seamless systems and featured for integrated microneedle-based theranostic systems, which serve as a bridge of real-time monitoring, on-demand drug delivery, advanced electronic engineering, and data science for personalized disease management and remote medical application. Additionally, we discuss the challenges and prospects of integrated microneedle-based theranostic systems for precision medicine and clinical application.
Collapse
Affiliation(s)
- Yiming Qin
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing 400038, China; Department of Dermatology and Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Feiyun Cui
- School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, China
| | - Yifei Lu
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Peng Yang
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Weiming Gou
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Zixuan Tang
- School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, China
| | - Shan Lu
- School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, China
| | - H Susan Zhou
- Department of Chemical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA 01609, United States
| | - Gaoxing Luo
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.
| | - Xiaoyan Lyu
- Department of Dermatology and Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Qing Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
15
|
Kretschmer F, Seipp J, Ludwig M, Klau GW, Böcker S. Coverage bias in small molecule machine learning. Nat Commun 2025; 16:554. [PMID: 39788952 PMCID: PMC11718084 DOI: 10.1038/s41467-024-55462-w] [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: 09/11/2023] [Accepted: 12/12/2024] [Indexed: 01/12/2025] Open
Abstract
Small molecule machine learning aims to predict chemical, biochemical, or biological properties from molecular structures, with applications such as toxicity prediction, ligand binding, and pharmacokinetics. A recent trend is developing end-to-end models that avoid explicit domain knowledge. These models assume no coverage bias in training and evaluation data, meaning the data are representative of the true distribution. However, the domain of applicability is rarely considered in such models. Here, we investigate how well large-scale datasets cover the space of known biomolecular structures. For doing so, we propose a distance measure based on solving the Maximum Common Edge Subgraph (MCES) problem, which aligns well with chemical similarity. Although this method is computationally hard, we introduce an efficient approach combining Integer Linear Programming and heuristic bounds. Our findings reveal that many widely-used datasets lack uniform coverage of biomolecular structures, limiting the predictive power of models trained on them. We propose two additional methods to assess whether training datasets diverge from known molecular distributions, potentially guiding future dataset creation to improve model performance.
Collapse
Affiliation(s)
- Fleming Kretschmer
- Chair for Bioinformatics, Institute for Computer Science, Friedrich Schiller University Jena, Jena, Germany
| | - Jan Seipp
- Algorithmic Bioinformatics, Institute for Computer Science, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Marcus Ludwig
- Chair for Bioinformatics, Institute for Computer Science, Friedrich Schiller University Jena, Jena, Germany
- Currently at Bright Giant, Jena, Germany
| | - Gunnar W Klau
- Algorithmic Bioinformatics, Institute for Computer Science, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sebastian Böcker
- Chair for Bioinformatics, Institute for Computer Science, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
16
|
Wilhelm C, Steckelberg A, Rebitschek FG. Benefits and harms associated with the use of AI-related algorithmic decision-making systems by healthcare professionals: a systematic review. THE LANCET REGIONAL HEALTH. EUROPE 2025; 48:101145. [PMID: 39687669 PMCID: PMC11648885 DOI: 10.1016/j.lanepe.2024.101145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 12/18/2024]
Abstract
Background Despite notable advancements in artificial intelligence (AI) that enable complex systems to perform certain tasks more accurately than medical experts, the impact on patient-relevant outcomes remains uncertain. To address this gap, this systematic review assesses the benefits and harms associated with AI-related algorithmic decision-making (ADM) systems used by healthcare professionals, compared to standard care. Methods In accordance with the PRISMA guidelines, we included interventional and observational studies published as peer-reviewed full-text articles that met the following criteria: human patients; interventions involving algorithmic decision-making systems, developed with and/or utilizing machine learning (ML); and outcomes describing patient-relevant benefits and harms that directly affect health and quality of life, such as mortality and morbidity. Studies that did not undergo preregistration, lacked a standard-of-care control, or pertained to systems that assist in the execution of actions (e.g., in robotics) were excluded. We searched MEDLINE, EMBASE, IEEE Xplore, and Google Scholar for studies published in the past decade up to 31 March 2024. We assessed risk of bias using Cochrane's RoB 2 and ROBINS-I tools, and reporting transparency with CONSORT-AI and TRIPOD-AI. Two researchers independently managed the processes and resolved conflicts through discussion. This review has been registered with PROSPERO (CRD42023412156) and the study protocol has been published. Findings Out of 2,582 records identified after deduplication, 18 randomized controlled trials (RCTs) and one cohort study met the inclusion criteria, covering specialties such as psychiatry, oncology, and internal medicine. Collectively, the studies included a median of 243 patients (IQR 124-828), with a median of 50.5% female participants (range 12.5-79.0, IQR 43.6-53.6) across intervention and control groups. Four studies were classified as having low risk of bias, seven showed some concerns, and another seven were assessed as having high or serious risk of bias. Reporting transparency varied considerably: six studies showed high compliance, four moderate, and five low compliance with CONSORT-AI or TRIPOD-AI. Twelve studies (63%) reported patient-relevant benefits. Of those with low risk of bias, interventions reduced length of stay in hospital and intensive care unit (10.3 vs. 13.0 days, p = 0.042; 6.3 vs. 8.4 days, p = 0.030), in-hospital mortality (9.0% vs. 21.3%, p = 0.018), and depression symptoms in non-complex cases (45.1% vs. 52.3%, p = 0.03). However, harms were frequently underreported, with only eight studies (42%) documenting adverse events. No study reported an increase in adverse events as a result of the interventions. Interpretation The current evidence on AI-related ADM systems provides limited insights into patient-relevant outcomes. Our findings underscore the essential need for rigorous evaluations of clinical benefits, reinforced compliance with methodological standards, and balanced consideration of both benefits and harms to ensure meaningful integration into healthcare practice. Funding This study did not receive any funding.
Collapse
Affiliation(s)
- Christoph Wilhelm
- International Graduate Academy (InGrA), Institute of Health and Nursing Science, Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 8, Halle (Saale) 06112, Germany
- Harding Center for Risk Literacy, Faculty of Health Sciences Brandenburg, University of Potsdam, Virchowstr. 2, Potsdam 14482, Germany
| | - Anke Steckelberg
- Institute of Health and Nursing Science, Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 8, Halle (Saale) 06112, Germany
| | - Felix G. Rebitschek
- Harding Center for Risk Literacy, Faculty of Health Sciences Brandenburg, University of Potsdam, Virchowstr. 2, Potsdam 14482, Germany
- Max Planck Institute for Human Development, Lentzeallee 94, Berlin 14195, Germany
| |
Collapse
|
17
|
Newman-Griffis D. AI Thinking: a framework for rethinking artificial intelligence in practice. ROYAL SOCIETY OPEN SCIENCE 2025; 12:241482. [PMID: 39780964 PMCID: PMC11706651 DOI: 10.1098/rsos.241482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025]
Abstract
Artificial intelligence is transforming the way we work with information across disciplines and practical contexts. A growing range of disciplines are now involved in studying, developing and assessing the use of AI in practice, but these disciplines often employ conflicting understandings of what AI is and what is involved in its use. New, interdisciplinary approaches are needed to bridge competing conceptualizations of AI in practice and help shape the future of AI use. I propose a novel conceptual framework called AI Thinking, which models key decisions and considerations involved in AI use across disciplinary perspectives. AI Thinking addresses five practice-based competencies involved in applying AI in context: motivating AI use, formulating AI methods, assessing available tools and technologies, selecting appropriate data and situating AI in the sociotechnical contexts it is used in. A hypothetical case study is provided to illustrate the application of AI Thinking in practice. This article situates AI Thinking in broader cross-disciplinary discourses of AI, including its connections to ongoing discussions around AI literacy and AI-driven innovation. AI Thinking can help to bridge between the work of diverse disciplines, contexts and actors in the AI space, and shape AI efforts in education, industrial development and policy.
Collapse
Affiliation(s)
- Denis Newman-Griffis
- Centre for Machine Intelligence, The University of Sheffield, SheffieldS1 3JD, UK
- Information School, The University of Sheffield, SheffieldS10 2AH, UK
- Research on Research Institute, LondonWC1E 6JA, UK
| |
Collapse
|
18
|
Santos ESA, Lemos JM, dos Santos Carvalho AM, Mendonça de Melo FDS, Pereira EDS, Moreira-Filho JT, Braga RDC, Muratov EN, Grellier P, Charneau S, Bastos IM, Neves BJ. Deep Multitask Learning-Driven Discovery of New Compounds Targeting Leishmania infantum. ACS OMEGA 2024; 9:51271-51284. [PMID: 39758621 PMCID: PMC11696749 DOI: 10.1021/acsomega.4c07994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 01/07/2025]
Abstract
Visceral leishmaniasis caused by Leishmania infantum is a severe and often fatal disease prevalent in low- and middle-income countries. Existing treatments are hampered by toxicity, high costs, and the emergence of drug resistance, highlighting the urgent need for novel therapeutics. In this context, we developed an explainable multitask learning (MTL) pipeline to predict the antileishmanial activity of compounds against three Leishmania species, with a primary focus on L. infantum. Then, we screened ∼1.3 million compounds from the ChemBridge database by using these models. This approach identified 20 putative hits, with nine compounds demonstrating significant in vitro antileishmanial activity against L. infantum. Three compounds exhibited notable potencies (IC50 of 1.05-15.6 μM) and moderate cytotoxicities (CC50 of 32.4 to >175 μM), positioning them as promising candidates for further hit-to-lead optimization. Our study underscores the effectiveness of multitask learning models in virtual screening, enabling the discovery of potent and selective antileishmanial compounds targeting L. infantum. Incorporating explainable techniques offers critical insights into the structural determinants of biological activity, aiding in the rational design and optimization of new therapeutics. These findings advocate for the potential of multitask learning methodologies to enhance hit rates in drug discovery for neglected tropical diseases.
Collapse
Affiliation(s)
| | - Jade Milhomem Lemos
- Laboratory
of Cheminformatics, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia 74605-170, Brazil
| | - Alexandra Maria dos Santos Carvalho
- Pathogen-Host
Interface Laboratory, Department of Cell Biology, Institute of Biological
Sciences, Universidade de Brasilia, Brasilia 70910-900, Brazil
| | - Felipe da Silva Mendonça de Melo
- Pathogen-Host
Interface Laboratory, Department of Cell Biology, Institute of Biological
Sciences, Universidade de Brasilia, Brasilia 70910-900, Brazil
| | - Eufrasia de Sousa Pereira
- Laboratory
of Cheminformatics, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia 74605-170, Brazil
| | | | | | - Eugene N. Muratov
- Laboratory
for Molecular Modeling, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7360, United
States
| | - Philippe Grellier
- UMR
7245 Molécules de Communication et Adaptation des Micro-organismes,
Muséum National d’Histoire Naturelle, Équipe Parasites et Protistes
Libres, Paris 75005, France
| | - Sébastien Charneau
- Laboratory
of Protein Chemistry and Biochemistry, Department of Cell Biology,
Institute of Biological Sciences, Universidade
de Brasilia, Brasilia 70910-900, Brazil
| | - Izabela Marques
Dourado Bastos
- Pathogen-Host
Interface Laboratory, Department of Cell Biology, Institute of Biological
Sciences, Universidade de Brasilia, Brasilia 70910-900, Brazil
| | - Bruno Junior Neves
- Laboratory
of Cheminformatics, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia 74605-170, Brazil
| |
Collapse
|
19
|
Rasul A, Hossain MS, Dastider AG, Roy H, Hasan MZ, Khosru QDM. A machine learning based classifier for topological quantum materials. Sci Rep 2024; 14:31564. [PMID: 39738190 DOI: 10.1038/s41598-024-68920-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 07/30/2024] [Indexed: 01/01/2025] Open
Abstract
Prediction and discovery of new materials with desired properties are at the forefront of quantum science and technology research. A major bottleneck in this field is the computational resources and time complexity related to finding new materials from ab initio calculations. In this work, an effective and robust deep learning-based model is proposed by incorporating persistent homology with graph neural network which offers an accuracy of 91.4 % and an F1 score of 88.5 % in classifying topological versus non-topological materials, outperforming the other state-of-the-art classifier models. Additionally, out-of-distribution and newly discovered topological materials can be classified using our method with high confidence. The incorporation of the graph neural network encodes the underlying relation between the atoms into the model based on their crystalline structures and thus proved to be an effective method to represent and process non-Euclidean data like molecules with a relatively shallow network. The persistent homology pipeline in the proposed neural network integrates a topological analysis of crystal structures into the deep learning model, enhancing both robustness and performance. Our classifier can serve as an efficacious tool for predicting the topological class, thereby enabling a high-throughput search for fascinating topological materials.
Collapse
Affiliation(s)
- Ashiqur Rasul
- Department of Electrical and Electronic Engineering, Bangladesh University of Engineering and Technology, Dhaka, 1205, Bangladesh.
| | | | - Ankan Ghosh Dastider
- Department of Electrical and Electronic Engineering, Bangladesh University of Engineering and Technology, Dhaka, 1205, Bangladesh
| | - Himaddri Roy
- Department of Electrical and Electronic Engineering, Bangladesh University of Engineering and Technology, Dhaka, 1205, Bangladesh
| | - M Zahid Hasan
- Department of Physics, Princeton University, Princeton, NJ, 08544, USA
| | - Quazi D M Khosru
- Department of Electrical and Electronic Engineering, Bangladesh University of Engineering and Technology, Dhaka, 1205, Bangladesh
| |
Collapse
|
20
|
Li X, Chen H, Yan J, Liu G, Li C, Zhou X, Wang Y, Wu Y, Yan B, Yan X. Balancing the Functionality and Biocompatibility of Materials with a Deep-Learning-Based Inverse Design Framework. ENVIRONMENT & HEALTH (WASHINGTON, D.C.) 2024; 2:875-885. [PMID: 39722843 PMCID: PMC11667291 DOI: 10.1021/envhealth.4c00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 12/28/2024]
Abstract
The rational design of molecules with the desired functionality presents a significant challenge in chemistry. Moreover, it is worth noting that making chemicals safe and sustainable is crucial to bringing them to the market. To address this, we propose a novel deep learning framework developed explicitly for inverse design of molecules with both functionality and biocompatibility. This innovative approach comprises two predictive models and one generative model, facilitating the targeted screening of novel molecules from created virtual chemical space. Our method's versatility is highlighted in the inverse design process, where it successfully generates molecules with specified motifs or composition, discovers synthetically accessible molecules, and jointly targets functional and safe properties beyond the training regime. The utility of this method is demonstrated in its ability to design ionic liquids (ILs) with enhanced antibacterial properties and reduced cytotoxicity, addressing the issue of balancing functionality and biocompatibility in molecular design.
Collapse
Affiliation(s)
- Xiaofang Li
- Institute
of Environmental Research at Greater Bay Area, Key Laboratory for
Water Quality and Conservation of the Pearl River Delta, Ministry
of Education, Guangzhou University, Guangzhou 510006, China
| | - Hanle Chen
- Institute
of Environmental Research at Greater Bay Area, Key Laboratory for
Water Quality and Conservation of the Pearl River Delta, Ministry
of Education, Guangzhou University, Guangzhou 510006, China
| | - Jiachen Yan
- Institute
of Environmental Research at Greater Bay Area, Key Laboratory for
Water Quality and Conservation of the Pearl River Delta, Ministry
of Education, Guangzhou University, Guangzhou 510006, China
| | - Guohong Liu
- School
of Health, Guangzhou Vocational University
of Science and Technology, Guangzhou 510555, China
| | - Chengjun Li
- Institute
of Environmental Research at Greater Bay Area, Key Laboratory for
Water Quality and Conservation of the Pearl River Delta, Ministry
of Education, Guangzhou University, Guangzhou 510006, China
| | - Xiaoxia Zhou
- Institute
of Environmental Research at Greater Bay Area, Key Laboratory for
Water Quality and Conservation of the Pearl River Delta, Ministry
of Education, Guangzhou University, Guangzhou 510006, China
| | - Yan Wang
- College
of Animal Science, South China Agricultural
University, Guangzhou 510642, China
| | - Yinbao Wu
- College
of Animal Science, South China Agricultural
University, Guangzhou 510642, China
| | - Bing Yan
- Institute
of Environmental Research at Greater Bay Area, Key Laboratory for
Water Quality and Conservation of the Pearl River Delta, Ministry
of Education, Guangzhou University, Guangzhou 510006, China
| | - Xiliang Yan
- Institute
of Environmental Research at Greater Bay Area, Key Laboratory for
Water Quality and Conservation of the Pearl River Delta, Ministry
of Education, Guangzhou University, Guangzhou 510006, China
- College
of Animal Science, South China Agricultural
University, Guangzhou 510642, China
| |
Collapse
|
21
|
Shahzadi M, Rafique H, Waheed A, Naz H, Waheed A, Zokirova FR, Khan H. Artificial intelligence for chimeric antigen receptor-based therapies: a comprehensive review of current applications and future perspectives. Ther Adv Vaccines Immunother 2024; 12:25151355241305856. [PMID: 39691280 PMCID: PMC11650588 DOI: 10.1177/25151355241305856] [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: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
Using artificial intelligence (AI) to enhance chimeric antigen receptor (CAR)-based therapies' design, production, and delivery is a novel and promising approach. This review provides an overview of the current applications and challenges of AI for CAR-based therapies and suggests some directions for future research and development. This paper examines some of the recent advances of AI for CAR-based therapies, for example, using deep learning (DL) to design CARs that target multiple antigens and avoid antigen escape; using natural language processing to extract relevant information from clinical reports and literature; using computer vision to analyze the morphology and phenotype of CAR cells; using reinforcement learning to optimize the dose and schedule of CAR infusion; and using AI to predict the efficacy and toxicity of CAR-based therapies. These applications demonstrate the potential of AI to improve the quality and efficiency of CAR-based therapies and to provide personalized and precise treatments for cancer patients. However, there are also some challenges and limitations of using AI for CAR-based therapies, for example, the lack of high-quality and standardized data; the need for validation and verification of AI models; the risk of bias and error in AI outputs; the ethical, legal, and social issues of using AI for health care; and the possible impact of AI on the human role and responsibility in cancer immunotherapy. It is important to establish a multidisciplinary collaboration among researchers, clinicians, regulators, and patients to address these challenges and to ensure the safe and responsible use of AI for CAR-based therapies.
Collapse
Affiliation(s)
- Muqadas Shahzadi
- Department of Zoology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | - Hamad Rafique
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Ahmad Waheed
- Department of Zoology, Faculty of Life Sciences, University of Okara, 2 KM Lahore Road, Renala Khurd, Okara 56130, Punjab, Pakistan
| | - Hina Naz
- Department of Zoology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | - Atifa Waheed
- Department of Biology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | | | - Humera Khan
- Department of Biochemistry, Sahiwal Medical College, Sahiwal, Pakistan
| |
Collapse
|
22
|
Liu Y, Su X, Ding J, Zhou J, Liu Z, Wei X, Yang HB, Liu B. Progress and challenges in structural, in situ and operando characterization of single-atom catalysts by X-ray based synchrotron radiation techniques. Chem Soc Rev 2024; 53:11850-11887. [PMID: 39434695 DOI: 10.1039/d3cs00967j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Single-atom catalysts (SACs) represent the ultimate size limit of nanoscale catalysts, combining the advantages of homogeneous and heterogeneous catalysts. SACs have isolated single-atom active sites that exhibit high atomic utilization efficiency, unique catalytic activity, and selectivity. Over the past few decades, synchrotron radiation techniques have played a crucial role in studying single-atom catalysis by identifying catalyst structures and enabling the understanding of reaction mechanisms. The profound comprehension of spectroscopic techniques and characteristics pertaining to SACs is important for exploring their catalytic activity origins and devising high-performance and stable SACs for industrial applications. In this review, we provide a comprehensive overview of the recent advances in X-ray based synchrotron radiation techniques for structural characterization and in situ/operando observation of SACs under reaction conditions. We emphasize the correlation between spectral fine features and structural characteristics of SACs, along with their analytical limitations. The development of IMST with spatial and temporal resolution is also discussed along with their significance in revealing the structural characteristics and reaction mechanisms of SACs. Additionally, this review explores the study of active center states using spectral fine characteristics combined with theoretical simulations, as well as spectroscopic analysis strategies utilizing machine learning methods to address challenges posed by atomic distribution inhomogeneity in SACs while envisaging potential applications integrating artificial intelligence seamlessly with experiments for real-time monitoring of single-atom catalytic processes.
Collapse
Affiliation(s)
- Yuhang Liu
- Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China.
- School of Materials Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China.
| | - Xiaozhi Su
- Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China.
| | - Jie Ding
- Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong SAR 999077, China.
| | - Jing Zhou
- College of Physics and Electronic Information Engineering, Zhejiang Normal University, Jinhua 321004, China
| | - Zhen Liu
- Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China.
| | - Xiangjun Wei
- Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China.
| | - Hong Bin Yang
- School of Materials Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China.
| | - Bin Liu
- Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong SAR 999077, China.
- Department of Chemistry, Hong Kong Institute of Clean Energy (HKICE) & Center of Super-Diamond and Advanced Films (COSDAF), City University of Hong Kong, Hong Kong SAR 999077, China
| |
Collapse
|
23
|
Rao L, Yuan Y, Shen X, Yu G, Chen X. Designing nanotheranostics with machine learning. NATURE NANOTECHNOLOGY 2024; 19:1769-1781. [PMID: 39362960 DOI: 10.1038/s41565-024-01753-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/08/2024] [Indexed: 10/05/2024]
Abstract
The inherent limits of traditional diagnoses and therapies have driven the development and application of emerging nanotechnologies for more effective and safer management of diseases, herein referred to as 'nanotheranostics'. Although many important technological successes have been achieved in this field, widespread adoption of nanotheranostics as a new paradigm is hindered by specific obstacles, including time-consuming synthesis of nanoparticles, incomplete understanding of nano-bio interactions, and challenges regarding chemistry, manufacturing and the controls required for clinical translation and commercialization. As a key branch of artificial intelligence, machine learning (ML) provides a set of tools capable of performing time-consuming and result-perception tasks, thus offering unique opportunities for nanotheranostics. This Review summarizes the progress and challenges in this emerging field of ML-aided nanotheranostics, and discusses the opportunities in developing next-generation nanotheranostics with reliable datasets and advanced ML models to offer better clinical benefits to patients.
Collapse
Affiliation(s)
- Lang Rao
- Shenzhen Bay Laboratory, Shenzhen, China.
| | - Yuan Yuan
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Computer Science, Boston College, Chestnut Hill, MA, USA
| | - Xi Shen
- Tencent AI Lab, Shenzhen, China
- Intellindust, Shenzhen, China
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| |
Collapse
|
24
|
Heydari S, Masoumi N, Esmaeeli E, Ayyoubzadeh SM, Ghorbani-Bidkorpeh F, Ahmadi M. Artificial intelligence in nanotechnology for treatment of diseases. J Drug Target 2024; 32:1247-1266. [PMID: 39155708 DOI: 10.1080/1061186x.2024.2393417] [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: 04/17/2024] [Revised: 07/06/2024] [Accepted: 08/11/2024] [Indexed: 08/20/2024]
Abstract
Nano-based drug delivery systems (DDSs) have demonstrated the ability to address challenges posed by therapeutic agents, enhancing drug efficiency and reducing side effects. Various nanoparticles (NPs) are utilised as DDSs with unique characteristics, leading to diverse applications across different diseases. However, the complexity, cost and time-consuming nature of laboratory processes, the large volume of data, and the challenges in data analysis have prompted the integration of artificial intelligence (AI) tools. AI has been employed in designing, characterising and manufacturing drug delivery nanosystems, as well as in predicting treatment efficiency. AI's potential to personalise drug delivery based on individual patient factors, optimise formulation design and predict drug properties has been highlighted. By leveraging AI and large datasets, developing safe and effective DDSs can be accelerated, ultimately improving patient outcomes and advancing pharmaceutical sciences. This review article investigates the role of AI in the development of nano-DDSs, with a focus on their therapeutic applications. The use of AI in DDSs has the potential to revolutionise treatment optimisation and improve patient care.
Collapse
Affiliation(s)
- Soroush Heydari
- Department of Health Information Management, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloofar Masoumi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Erfan Esmaeeli
- Department of Health Information Management, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Ayyoubzadeh
- Department of Health Information Management, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
- Health Information Management Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ghorbani-Bidkorpeh
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahnaz Ahmadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Puhl AC, Lewicki SA, Gao ZG, Pramanik A, Makarov V, Ekins S, Jacobson KA. Machine learning-aided search for ligands of P2Y 6 and other P2Y receptors. Purinergic Signal 2024; 20:617-627. [PMID: 38526670 PMCID: PMC11554998 DOI: 10.1007/s11302-024-10003-4] [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: 01/24/2024] [Accepted: 03/12/2024] [Indexed: 03/27/2024] Open
Abstract
The P2Y6 receptor, activated by uridine diphosphate (UDP), is a target for antagonists in inflammatory, neurodegenerative, and metabolic disorders, yet few potent and selective antagonists are known to date. This prompted us to use machine learning as a novel approach to aid ligand discovery, with pharmacological evaluation at three P2YR subtypes: initially P2Y6 and subsequently P2Y1 and P2Y14. Relying on extensive published data for P2Y6R agonists, we generated and validated an array of classification machine learning model using the algorithms deep learning (DL), adaboost classifier (ada), Bernoulli NB (bnb), k-nearest neighbors (kNN) classifier, logistic regression (lreg), random forest classifier (rf), support vector classification (SVC), and XGBoost (XGB) classifier models, and the common consensus was applied to molecular selection of 21 diverse structures. Compounds were screened using human P2Y6R-induced functional calcium transients in transfected 1321N1 astrocytoma cells and fluorescent binding inhibition at closely related hP2Y14R expressed in CHO cells. The hit compound ABBV-744, an experimental anticancer drug with a 6-methyl-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyridine scaffold, had multifaceted interactions with the P2YR family: hP2Y6R inhibition in a non-surmountable fashion, suggesting that noncompetitive antagonism, and hP2Y1R enhancement, but not hP2Y14R binding inhibition. Other machine learning-selected compounds were either weak (experimental anti-asthmatic drug AZD5423 with a phenyl-1H-indazole scaffold) or inactive in inhibiting the hP2Y6R. Experimental drugs TAK-593 and GSK1070916 (100 µM) inhibited P2Y14R fluorescent binding by 50% and 38%, respectively, and all other compounds by < 20%. Thus, machine learning has led the way toward revealing previously unknown modulators of several P2YR subtypes that have varied effects.
Collapse
Affiliation(s)
- Ana C Puhl
- Collaborations Pharmaceuticals, Inc, 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Sarah A Lewicki
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Asmita Pramanik
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Vadim Makarov
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071, Moscow, Russian Federation
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc, 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA.
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
26
|
Hashemi S, Vosough P, Taghizadeh S, Savardashtaki A. Therapeutic peptide development revolutionized: Harnessing the power of artificial intelligence for drug discovery. Heliyon 2024; 10:e40265. [PMID: 39605829 PMCID: PMC11600032 DOI: 10.1016/j.heliyon.2024.e40265] [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: 02/20/2024] [Revised: 10/07/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Due to the spread of antibiotic resistance, global attention is focused on its inhibition and the expansion of effective medicinal compounds. The novel functional properties of peptides have opened up new horizons in personalized medicine. With artificial intelligence methods combined with therapeutic peptide products, pharmaceuticals and biotechnology advance drug development rapidly and reduce costs. Short-chain peptides inhibit a wide range of pathogens and have great potential for targeting diseases. To address the challenges of synthesis and sustainability, artificial intelligence methods, namely machine learning, must be integrated into their production. Learning methods can use complicated computations to select the active and toxic compounds of the drug and its metabolic activity. Through this comprehensive review, we investigated the artificial intelligence method as a potential tool for finding peptide-based drugs and providing a more accurate analysis of peptides through the introduction of predictable databases for effective selection and development.
Collapse
Affiliation(s)
- Samaneh Hashemi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parisa Vosough
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Taghizadeh
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Science Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
27
|
Liu F, Xu H, Cui P, Li S, Wang H, Wu Z. NFSA-DTI: A Novel Drug-Target Interaction Prediction Model Using Neural Fingerprint and Self-Attention Mechanism. Int J Mol Sci 2024; 25:11818. [PMID: 39519369 PMCID: PMC11546351 DOI: 10.3390/ijms252111818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/24/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Existing deep learning methods have shown outstanding performance in predicting drug-target interactions. However, they still have limitations: (1) the over-reliance on locally extracted features by some single encoders, with insufficient consideration of global features, and (2) the inadequate modeling and learning of local crucial interaction sites in drug-target interaction pairs. In this study, we propose a novel drug-target interaction prediction model called the Neural Fingerprint and Self-Attention Mechanism (NFSA-DTI), which effectively integrates the local information of drug molecules and target sequences with their respective global features. The neural fingerprint method is used in this model to extract global features of drug molecules, while the self-attention mechanism is utilized to enhance CNN's capability in capturing the long-distance dependencies between the subsequences in the target amino acid sequence. In the feature fusion module, we improve the bilinear attention network by incorporating attention pooling, which enhances the model's ability to learn local crucial interaction sites in the drug-target pair. The experimental results on three benchmark datasets demonstrated that NFSA-DTI outperformed all baseline models in predictive performance. Furthermore, case studies illustrated that our model could provide valuable insights for drug discovery. Moreover, our model offers molecular-level interpretations.
Collapse
Affiliation(s)
| | | | - Peng Cui
- School of Information, Guizhou University of Finance and Economics, Guiyang 550025, China; (F.L.)
| | | | | | - Ziye Wu
- School of Information, Guizhou University of Finance and Economics, Guiyang 550025, China; (F.L.)
| |
Collapse
|
28
|
Shao X, Pham A, Kuo TT. WebQuorumChain: A web framework for quorum-based health care model learning. INFORMATICS IN MEDICINE UNLOCKED 2024; 50:101590. [PMID: 39483487 PMCID: PMC11526443 DOI: 10.1016/j.imu.2024.101590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024] Open
Abstract
Background Institutions interested in collaborative machine learning to enhance healthcare may be deterred by privacy concerns. Decentralized federated learning is a privacy-preserving and security-robust tool to promote cross-institutional learning, however, such frameworks require complex setups and advanced technical expertise. Here, we aim to improve their utilization by offering an intuitive, user-friendly, and secure system that integrates both front-end and back-end functionalities. Method We develop WebQuorumChain, an integrated system built upon the QuorumChain schema. We test the system on a 2-site network using two publicly available health datasets and measure the average vertical and horizontal-ensemble AUCs per dataset across 30 trials, as well as the average execution time of the system. Results Our system achieved consistently high AUCs for each dataset (0.94-0.96), with reasonable total execution times ranging from 5 to 20 min, inclusive of modeling and all other system overheads. The front-end displays event logs generated from back-end layers in real time, in sync with the progress of the underlying algorithm. Conclusions We develop a web-based system that supplies users with visual tools to configure the federated learning network, manage training sessions, and inspect the learning process. WebQuorumChain helps schedule and monitor low-level processes without violating the fundamental security promises of cross-institutional decentralized machine learning. The system also maintains predictive accuracy and runtime efficiency in the presence of additional layers. WebQuorumChain will help promote meaningful collaboration among healthcare institutions, who can retain full control of their data privacy while contributing to data-driven discoveries.
Collapse
Affiliation(s)
- Xiyan Shao
- UCSD Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
| | - Anh Pham
- UCSD Health Department of Biomedical Informatics, University of California San Diego, La Jolla, CA, USA
| | - Tsung-Ting Kuo
- UCSD Health Department of Biomedical Informatics, University of California San Diego, La Jolla, CA, USA
- Department of Biomedical Informatics and Data Science, School of Medicine, Yale University, New Haven, CT, USA
- Department of Surgery, School of Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
29
|
Yang Z, Shi A, Zhang R, Ji Z, Li J, Lyu J, Qian J, Chen T, Wang X, You F, Xie J. When Metal Nanoclusters Meet Smart Synthesis. ACS NANO 2024; 18:27138-27166. [PMID: 39316700 DOI: 10.1021/acsnano.4c09597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Atomically precise metal nanoclusters (MNCs) represent a fascinating class of ultrasmall nanoparticles with molecule-like properties, bridging conventional metal-ligand complexes and nanocrystals. Despite their potential for various applications, synthesis challenges such as a precise understanding of varied synthetic parameters and property-driven synthesis persist, hindering their full exploitation and wider application. Incorporating smart synthesis methodologies, including a closed-loop framework of automation, data interpretation, and feedback from AI, offers promising solutions to address these challenges. In this perspective, we summarize the closed-loop smart synthesis that has been demonstrated in various nanomaterials and explore the research frontiers of smart synthesis for MNCs. Moreover, the perspectives on the inherent challenges and opportunities of smart synthesis for MNCs are discussed, aiming to provide insights and directions for future advancements in this emerging field of AI for Science, while the integration of deep learning algorithms stands to substantially enrich research in smart synthesis by offering enhanced predictive capabilities, optimization strategies, and control mechanisms, thereby extending the potential of MNC synthesis.
Collapse
Affiliation(s)
- Zhucheng Yang
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Fuzhou 350207, P. R. China
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore 117585, Singapore
| | - Anye Shi
- Systems Engineering, College of Engineering, Cornell University, Ithaca, New York 14583, United States
| | - Ruixuan Zhang
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Fuzhou 350207, P. R. China
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore 117585, Singapore
| | - Zuowei Ji
- School of Humanities and Social Sciences, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, P. R. China
| | - Jiali Li
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Jingkuan Lyu
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Fuzhou 350207, P. R. China
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore 117585, Singapore
| | - Jing Qian
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Fuzhou 350207, P. R. China
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore 117585, Singapore
| | - Tiankai Chen
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, P. R. China
| | - Xiaonan Wang
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Fengqi You
- Systems Engineering, College of Engineering, Cornell University, Ithaca, New York 14583, United States
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
- Cornell University AI for Science Institute (CUAISci), Cornell University, Ithaca, New York 14853, United States
| | - Jianping Xie
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Fuzhou 350207, P. R. China
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore 117585, Singapore
| |
Collapse
|
30
|
Yarahmadi P, Ahmadpour E, Moradi P, Samadzadehaghdam N. Automatic classification of Giardia infection from stool microscopic images using deep neural networks. BIOIMPACTS : BI 2024; 15:30272. [PMID: 40161947 PMCID: PMC11954735 DOI: 10.34172/bi.30272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/23/2024] [Accepted: 05/28/2024] [Indexed: 04/02/2025]
Abstract
Introduction Giardiasis is a common intestinal infection caused by the Giardia lamblia parasite, and its rapid and accurate diagnosis is crucial for effective treatment. The automatic classification of Giardia infection from stool microscopic images plays a vital role in this diagnosis process. In this study, we applied deep learning-based models to automatically classify stool microscopic images into three categories, namely, normal, cyst, and trophozoite. Methods Unlike previous studies focused on images acquired from drinking water samples, we specifically targeted stool samples. In this regard, we collected a dataset of 1610 microscopic digital images captured by a smartphone with a resolution of 2340 × 1080 pixels from stool samples under the Nikon YS100 microscope. First, we applied CLAHE (Contrast Limited Adaptive Histogram Equalization) histogram equalization a method to enhance the image quality and contrast. We employed three deep learning models, namely Xception, ResNet-50, and EfficientNet-B0, to evaluate their classification performance. Each model was trained on the dataset of microscopic images and fine-tuned using transfer learning techniques. Results Among the three deep learning models, EfficientNet-B0 demonstrated superior performance in classifying Giardia lamblia parasites from stool microscopic images. The model achieved precision, accuracy, recall, specificity, and F1-score values of 0.9599, 0.9629, 0.9619, 0.9821, and 0.9607, respectively. Conclusion The EfficientNet-B0 showed promising results in accurately identifying normal, cyst, and trophozoite forms of Giardia lamblia parasites. This automated classification approach can provide valuable assistance to laboratory science experts and parasitologists in the rapid and accurate diagnosis of giardiasis, ultimately improving patient care and treatment outcomes.
Collapse
Affiliation(s)
- Pezhman Yarahmadi
- Department of Biomedical Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences Tabriz, Iran
| | - Ehsan Ahmadpour
- Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parham Moradi
- Department of Computer Engineering, University of Kurdistan, Sanandaj, Iran
| | - Nasser Samadzadehaghdam
- Department of Biomedical Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences Tabriz, Iran
| |
Collapse
|
31
|
Kokudeva M, Vichev M, Naseva E, Miteva DG, Velikova T. Artificial intelligence as a tool in drug discovery and development. World J Exp Med 2024; 14:96042. [PMID: 39312699 PMCID: PMC11372739 DOI: 10.5493/wjem.v14.i3.96042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/06/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024] Open
Abstract
The rapidly advancing field of artificial intelligence (AI) has garnered substantial attention for its potential application in drug discovery and development. This opinion review critically examined the feasibility and prospects of integrating AI as a transformative tool in the pharmaceutical industry. AI, encompassing machine learning algorithms, deep learning, and data analytics, offers unprecedented opportunities to streamline and enhance various stages of drug development. This opinion review delved into the current landscape of AI-driven approaches, discussing their utilization in target identification, lead optimization, and predictive modeling of pharmacokinetics and toxicity. We aimed to scrutinize the integration of large-scale omics data, electronic health records, and chemical informatics, highlighting the power of AI in uncovering novel therapeutic targets and accelerating drug repurposing strategies. Despite the considerable potential of AI, the review also addressed inherent challenges, including data privacy concerns, interpretability of AI models, and the need for robust validation in real-world clinical settings. Additionally, we explored ethical considerations surrounding AI-driven decision-making in drug development. This opinion review provided a nuanced perspective on the transformative role of AI in drug discovery by discussing the existing literature and emerging trends, presenting critical insights and addressing potential hurdles. In conclusion, this study aimed to stimulate discourse within the scientific community and guide future endeavors to harness the full potential of AI in drug development.
Collapse
Affiliation(s)
- Maria Kokudeva
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia 1000, Bulgaria
| | | | - Emilia Naseva
- Faculty of Public Health, Medical University of Sofia, Sofia 1431, Bulgaria
| | - Dimitrina Georgieva Miteva
- Department of Genetics, Faculty of Biology, Sofia University St. Kliment Ohridski, Sofia 1164, Bulgaria
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| |
Collapse
|
32
|
Mag P, Nemes-Terényi M, Jerzsele Á, Mátyus P. Some Aspects and Convergence of Human and Veterinary Drug Repositioning. Molecules 2024; 29:4475. [PMID: 39339469 PMCID: PMC11433938 DOI: 10.3390/molecules29184475] [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: 07/30/2024] [Revised: 09/11/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Drug innovation traditionally follows a de novo approach with new molecules through a complex preclinical and clinical pathway. In addition to this strategy, drug repositioning has also become an important complementary approach, which can be shorter, cheaper, and less risky. This review provides an overview of drug innovation in both human and veterinary medicine, with a focus on drug repositioning. The evolution of drug repositioning and the effectiveness of this approach are presented, including the growing role of data science and computational modeling methods in identifying drugs with potential for repositioning. Certain business aspects of drug innovation, especially the relevant factors of market exclusivity, are also discussed. Despite the promising potential of drug repositioning for innovation, it remains underutilized, especially in veterinary applications. To change this landscape for mutual benefits of human and veterinary drug innovation, further exploitation of the potency of drug repositioning is necessary through closer cooperation between all stakeholders, academia, industry, pharmaceutical authorities, and innovation policy makers, and the integration of human and veterinary repositioning into a unified innovation space. For this purpose, the establishment of the conceptually new "One Health Drug Repositioning Platform" is proposed. Oncology is one of the disease areas where this platform can significantly support the development of new drugs for human and dog (or other companion animals) anticancer therapies. As an example of the utilization of human and veterinary drugs for veterinary repositioning, the use of COX inhibitors to treat dog cancers is reviewed.
Collapse
Affiliation(s)
- Patrik Mag
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István Street 2, 1078 Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, István Street 2, 1078 Budapest, Hungary
| | - Melinda Nemes-Terényi
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István Street 2, 1078 Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, István Street 2, 1078 Budapest, Hungary
| | - Ákos Jerzsele
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István Street 2, 1078 Budapest, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, István Street 2, 1078 Budapest, Hungary
| | - Péter Mátyus
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, István Street 2, 1078 Budapest, Hungary
| |
Collapse
|
33
|
Zheng JJ, Li QZ, Wang Z, Wang X, Zhao Y, Gao X. Computer-aided nanodrug discovery: recent progress and future prospects. Chem Soc Rev 2024; 53:9059-9132. [PMID: 39148378 DOI: 10.1039/d3cs00575e] [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: 08/17/2024]
Abstract
Nanodrugs, which utilise nanomaterials in disease prevention and therapy, have attracted considerable interest since their initial conceptualisation in the 1990s. Substantial efforts have been made to develop nanodrugs for overcoming the limitations of conventional drugs, such as low targeting efficacy, high dosage and toxicity, and potential drug resistance. Despite the significant progress that has been made in nanodrug discovery, the precise design or screening of nanomaterials with desired biomedical functions prior to experimentation remains a significant challenge. This is particularly the case with regard to personalised precision nanodrugs, which require the simultaneous optimisation of the structures, compositions, and surface functionalities of nanodrugs. The development of powerful computer clusters and algorithms has made it possible to overcome this challenge through in silico methods, which provide a comprehensive understanding of the medical functions of nanodrugs in relation to their physicochemical properties. In addition, machine learning techniques have been widely employed in nanodrug research, significantly accelerating the understanding of bio-nano interactions and the development of nanodrugs. This review will present a summary of the computational advances in nanodrug discovery, focusing on the understanding of how the key interfacial interactions, namely, surface adsorption, supramolecular recognition, surface catalysis, and chemical conversion, affect the therapeutic efficacy of nanodrugs. Furthermore, this review will discuss the challenges and opportunities in computer-aided nanodrug discovery, with particular emphasis on the integrated "computation + machine learning + experimentation" strategy that can potentially accelerate the discovery of precision nanodrugs.
Collapse
Affiliation(s)
- Jia-Jia Zheng
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| | - Qiao-Zhi Li
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| | - Zhenzhen Wang
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| | - Xiaoli Wang
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Yuliang Zhao
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| | - Xingfa Gao
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| |
Collapse
|
34
|
Fatima A, Geethakumari AM, Ahmed WS, Biswas KH. A potential allosteric inhibitor of SARS-CoV-2 main protease (M pro) identified through metastable state analysis. Front Mol Biosci 2024; 11:1451280. [PMID: 39310374 PMCID: PMC11413593 DOI: 10.3389/fmolb.2024.1451280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/14/2024] [Indexed: 09/25/2024] Open
Abstract
Anti-COVID19 drugs, such as nirmatrelvir, have been developed targeting the SARS-CoV-2 main protease, Mpro, based on the critical requirement of its proteolytic processing of the viral polyproteins into functional proteins essential for viral replication. However, the emergence of SARS-CoV-2 variants with Mpro mutations has raised the possibility of developing resistance against these drugs, likely due to therapeutic targeting of the Mpro catalytic site. An alternative to these drugs is the development of drugs that target an allosteric site distant from the catalytic site in the protein that may reduce the chance of the emergence of resistant mutants. Here, we combine computational analysis with in vitro assay and report the discovery of a potential allosteric site and an allosteric inhibitor of SARS-CoV-2 Mpro. Specifically, we identified an Mpro metastable state with a deformed catalytic site harboring potential allosteric sites, raising the possibility that stabilization of this metastable state through ligand binding can lead to the inhibition of Mpro activity. We then performed a computational screening of a library (∼4.2 million) of drug-like compounds from the ZINC database and identified several candidate molecules with high predicted binding affinity. MD simulations showed stable binding of the three top-ranking compounds to the putative allosteric sites in the protein. Finally, we tested the three compounds in vitro using a BRET-based Mpro biosensor and found that one of the compounds (ZINC4497834) inhibited the Mpro activity. We envisage that the identification of a potential allosteric inhibitor of Mpro will aid in developing improved anti-COVID-19 therapy.
Collapse
|
35
|
Wilhelm C, Steckelberg A, Rebitschek FG. Is artificial intelligence for medical professionals serving the patients? : Protocol for a systematic review on patient-relevant benefits and harms of algorithmic decision-making. Syst Rev 2024; 13:228. [PMID: 39242544 PMCID: PMC11378383 DOI: 10.1186/s13643-024-02646-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/22/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Algorithmic decision-making (ADM) utilises algorithms to collect and process data and develop models to make or support decisions. Advances in artificial intelligence (AI) have led to the development of support systems that can be superior to medical professionals without AI support in certain tasks. However, whether patients can benefit from this remains unclear. The aim of this systematic review is to assess the current evidence on patient-relevant benefits and harms, such as improved survival rates and reduced treatment-related complications, when healthcare professionals use ADM systems (developed using or working with AI) compared to healthcare professionals without AI-related ADM (standard care)-regardless of the clinical issues. METHODS Following the PRISMA statement, MEDLINE and PubMed (via PubMed), Embase (via Elsevier) and IEEE Xplore will be searched using English free text terms in title/abstract, Medical Subject Headings (MeSH) terms and Embase Subject Headings (Emtree fields). Additional studies will be identified by contacting authors of included studies and through reference lists of included studies. Grey literature searches will be conducted in Google Scholar. Risk of bias will be assessed by using Cochrane's RoB 2 for randomised trials and ROBINS-I for non-randomised trials. Transparent reporting of the included studies will be assessed using the CONSORT-AI extension statement. Two researchers will screen, assess and extract from the studies independently, with a third in case of conflicts that cannot be resolved by discussion. DISCUSSION It is expected that there will be a substantial shortage of suitable studies that compare healthcare professionals with and without ADM systems concerning patient-relevant endpoints. This can be attributed to the prioritisation of technical quality criteria and, in some cases, clinical parameters over patient-relevant endpoints in the development of study designs. Furthermore, it is anticipated that a significant portion of the identified studies will exhibit relatively poor methodological quality and provide only limited generalisable results. SYSTEMATIC REVIEW REGISTRATION This study is registered within PROSPERO (CRD42023412156).
Collapse
Affiliation(s)
- Christoph Wilhelm
- Institute of Health and Nursing Sciences, Martin Luther University Halle-Wittenberg, Magdeburger Str. 8, Halle, 06112, Germany.
- Harding Center for Risk Literacy, Faculty of Health Sciences Brandenburg, University of Potsdam, Virchowstr. 2, Potsdam, 14482, Germany.
| | - Anke Steckelberg
- Institute of Health and Nursing Sciences, Martin Luther University Halle-Wittenberg, Magdeburger Str. 8, Halle, 06112, Germany
| | - Felix G Rebitschek
- Harding Center for Risk Literacy, Faculty of Health Sciences Brandenburg, University of Potsdam, Virchowstr. 2, Potsdam, 14482, Germany
- Max Planck Institute for Human Development, Lentzeallee 94, Berlin, 14195, Germany
| |
Collapse
|
36
|
Gharibshahian M, Torkashvand M, Bavisi M, Aldaghi N, Alizadeh A. Recent advances in artificial intelligent strategies for tissue engineering and regenerative medicine. Skin Res Technol 2024; 30:e70016. [PMID: 39189880 PMCID: PMC11348508 DOI: 10.1111/srt.70016] [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: 06/17/2024] [Accepted: 08/05/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Tissue engineering and regenerative medicine (TERM) aim to repair or replace damaged or lost tissues or organs due to accidents, diseases, or aging, by applying different sciences. For this purpose, an essential part of TERM is the designing, manufacturing, and evaluating of scaffolds, cells, tissues, and organs. Artificial intelligence (AI) or the intelligence of machines or software can be effective in all areas where computers play a role. METHODS The "artificial intelligence," "machine learning," "tissue engineering," "clinical evaluation," and "scaffold" keywords used for searching in various databases and articles published from 2000 to 2024 were evaluated. RESULTS The combination of tissue engineering and AI has created a new generation of technological advancement in the biomedical industry. Experience in TERM has been refined using advanced design and manufacturing techniques. Advances in AI, particularly deep learning, offer an opportunity to improve scientific understanding and clinical outcomes in TERM. CONCLUSION The findings of this research show the high potential of AI, machine learning, and robots in the selection, design, and fabrication of scaffolds, cells, tissues, or organs, and their analysis, characterization, and evaluation after their implantation. AI can be a tool to accelerate the introduction of tissue engineering products to the bedside. HIGHLIGHTS The capabilities of artificial intelligence (AI) can be used in different ways in all the different stages of TERM and not only solve the existing limitations, but also accelerate the processes, increase efficiency and precision, reduce costs, and complications after transplantation. ML predicts which technologies have the most efficient and easiest path to enter the market and clinic. The use of AI along with these imaging techniques can lead to the improvement of diagnostic information, the reduction of operator errors when reading images, and the improvement of image analysis (such as classification, localization, regression, and segmentation).
Collapse
Affiliation(s)
- Maliheh Gharibshahian
- Nervous System Stem Cells Research CenterSemnan University of Medical SciencesSemnanIran
- Department of Tissue Engineering and Applied Cell SciencesSchool of MedicineSemnan University of Medical SciencesSemnanIran
| | | | - Mahya Bavisi
- Department of Tissue Engineering and Applied Cell SciencesSchool of Advanced Technologies in MedicineIran University of Medical SciencesTehranIran
| | - Niloofar Aldaghi
- Student Research CommitteeSchool of MedicineShahroud University of Medical SciencesShahroudIran
| | - Akram Alizadeh
- Nervous System Stem Cells Research CenterSemnan University of Medical SciencesSemnanIran
- Department of Tissue Engineering and Applied Cell SciencesSchool of MedicineSemnan University of Medical SciencesSemnanIran
| |
Collapse
|
37
|
Ahmad F, Muhmood T. Clinical translation of nanomedicine with integrated digital medicine and machine learning interventions. Colloids Surf B Biointerfaces 2024; 241:114041. [PMID: 38897022 DOI: 10.1016/j.colsurfb.2024.114041] [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: 02/01/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024]
Abstract
Nanomaterials based therapeutics transform the ways of disease prevention, diagnosis and treatment with increasing sophistications in nanotechnology at a breakneck pace, but very few could reach to the clinic due to inconsistencies in preclinical studies followed by regulatory hinderances. To tackle this, integrating the nanomedicine discovery with digital medicine provide technologies as tools of specific biological activity measurement. Hence, overcome the redundancies in nanomedicine discovery by the on-site data acquisition and analytics through integrating intelligent sensors and artificial intelligence (AI) or machine learning (ML). Integrated AI/ML wearable sensors directly gather clinically relevant biochemical information from the subject's body and process data for physicians to make right clinical decision(s) in a time and cost-effective way. This review summarizes insights and recommend the infusion of actionable big data computation enabled sensors in burgeoning field of nanomedicine at academia, research institutes, and pharmaceutical industries, with a potential of clinical translation. Furthermore, many blind spots are present in modern clinically relevant computation, one of which could prevent ML-guided low-cost new nanomedicine development from being successfully translated into the clinic was also discussed.
Collapse
Affiliation(s)
- Farooq Ahmad
- State Key Laboratory of Chemistry and Utilization of Carbon Based Energy Resources, College of Chemistry, Xinjiang University, Urumqi 830017, China.
| | - Tahir Muhmood
- International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga, Braga 4715-330, Portugal.
| |
Collapse
|
38
|
Yeshi K, Jamtsho T, Wangchuk P. Current Treatments, Emerging Therapeutics, and Natural Remedies for Inflammatory Bowel Disease. Molecules 2024; 29:3954. [PMID: 39203033 PMCID: PMC11357616 DOI: 10.3390/molecules29163954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic, lifelong disorder characterized by inflammation of the gastrointestinal (GI) tract. The exact etiology of IBD remains incompletely understood due to its multifaceted nature, which includes genetic predisposition, environmental factors, and host immune response dysfunction. Currently, there is no cure for IBD. This review discusses the available treatment options and the challenges they present. Importantly, we examine emerging therapeutics, such as biologics and immunomodulators, that offer targeted treatment strategies for IBD. While many IBD patients do not respond adequately to most biologics, recent clinical trials combining biologics with small-molecule drugs (SMDs) have provided new insights into improving the IBD treatment landscape. Furthermore, numerous novel and specific therapeutic targets have been identified. The high cost of IBD drugs poses a significant barrier to treatment, but this challenge may be alleviated with the development of more affordable biosimilars. Additionally, emerging point-of-care protein biomarkers from serum and plasma are showing potential for enhancing the precision of IBD diagnosis and prognosis. Several natural products (NPs), including crude extracts, small molecules, and peptides, have demonstrated promising anti-inflammatory activity in high-throughput screening (HTS) systems and advanced artificial intelligence (AI)-assisted platforms, such as molecular docking and ADMET prediction. These platforms are advancing the search for alternative IBD therapies derived from natural sources, potentially leading to more affordable and safer treatment options with fewer side effects.
Collapse
Affiliation(s)
- Karma Yeshi
- College of Public Health, Medical, and Veterinary Sciences (CPHMVS), James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia;
- Australian Institute of Tropical Health and Medicine (AITHM), James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia
| | - Tenzin Jamtsho
- College of Public Health, Medical, and Veterinary Sciences (CPHMVS), James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia;
- Australian Institute of Tropical Health and Medicine (AITHM), James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia
| | - Phurpa Wangchuk
- College of Public Health, Medical, and Veterinary Sciences (CPHMVS), James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia;
- Australian Institute of Tropical Health and Medicine (AITHM), James Cook University, Building E4, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia
| |
Collapse
|
39
|
Dorsey MA, Dsouza K, Ranganath D, Harris JS, Lane TR, Urbina F, Ekins S. Near-Term Quantum Classification Algorithms Applied to Antimalarial Drug Discovery. J Chem Inf Model 2024; 64:5922-5930. [PMID: 39013438 PMCID: PMC11338495 DOI: 10.1021/acs.jcim.4c00953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Computational approaches are widely applied in drug discovery to explore properties related to bioactivity, physiochemistry, and toxicology. Over at least the last 20 years, the exploitation of machine learning on molecular data sets has been used to understand the structure-activity relationships that exist between biomolecules and druggable targets. More recently, these methods have also seen application for phenotypic screening data for neglected diseases such as tuberculosis and malaria. Herein, we apply machine learning to build quantum Quantitative Structure Activity Relationship models from antimalarial data sets. There is a continual need for new antimalarials to address drug resistance, and the readily available in vitro data sets could be utilized with newer machine learning approaches as these develop. Furthermore, quantum machine learning is a relatively new method that uses a quantum computer to perform the calculations. First, we present a classical-quantum hybrid computational approach by building a Latent Bernoulli Autoencoder machine learning model for compressing bit-vector descriptors to a size that can be adapted to quantum computers for classification tasks with limited loss of embedded information. Second, we apply our method for feature map compression to quantum classification algorithms, including a completely novel machine learning algorithm with no analogy in classical computers: the Quantum Fourier Transform Classifier. We apply both these approaches to build quantum machine learning models for small-molecule antimalarials with quantum simulation software and then benchmark these quantum models against classical machine learning approaches. While there are many challenges currently facing the development of reliable quantum computers, our results demonstrate that there is potential for the use of this technology in the field of drug discovery.
Collapse
Affiliation(s)
- Matthew A. Dorsey
- Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27606, United States
| | - Kelvin Dsouza
- Electrical and Computer Engineering, North Carolina State University, Raleigh, North Carolina 27606, United States
| | - Dhruv Ranganath
- Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | - Joshua S. Harris
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Thomas R. Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Fabio Urbina
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| |
Collapse
|
40
|
Catacutan DB, Alexander J, Arnold A, Stokes JM. Machine learning in preclinical drug discovery. Nat Chem Biol 2024:10.1038/s41589-024-01679-1. [PMID: 39030362 DOI: 10.1038/s41589-024-01679-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/13/2024] [Indexed: 07/21/2024]
Abstract
Drug-discovery and drug-development endeavors are laborious, costly and time consuming. These programs can take upward of 12 years and cost US $2.5 billion, with a failure rate of more than 90%. Machine learning (ML) presents an opportunity to improve the drug-discovery process. Indeed, with the growing abundance of public and private large-scale biological and chemical datasets, ML techniques are becoming well positioned as useful tools that can augment the traditional drug-development process. In this Perspective, we discuss the integration of algorithmic methods throughout the preclinical phases of drug discovery. Specifically, we highlight an array of ML-based efforts, across diverse disease areas, to accelerate initial hit discovery, mechanism-of-action (MOA) elucidation and chemical property optimization. With advances in the application of ML across diverse therapeutic areas, we posit that fully ML-integrated drug-discovery pipelines will define the future of drug-development programs.
Collapse
Affiliation(s)
- Denise B Catacutan
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
| | - Jeremie Alexander
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
| | - Autumn Arnold
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan M Stokes
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada.
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
41
|
Bai X, Xie Y, Zhang X, Han H, Li JR. Evaluation of Open-Source Large Language Models for Metal-Organic Frameworks Research. J Chem Inf Model 2024; 64:4958-4965. [PMID: 38529913 DOI: 10.1021/acs.jcim.4c00065] [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: 03/27/2024]
Abstract
Along with the development of machine learning, deep learning, and large language models (LLMs) such as GPT-4 (GPT: Generative Pre-Trained Transformer), artificial intelligence (AI) tools have been playing an increasingly important role in chemical and material research to facilitate the material screening and design. Despite the exciting progress of GPT-4 based AI research assistance, open-source LLMs have not gained much attention from the scientific community. This work primarily focused on metal-organic frameworks (MOFs) as a subdomain of chemistry and evaluated six top-rated open-source LLMs with a comprehensive set of tasks including MOFs knowledge, basic chemistry knowledge, in-depth chemistry knowledge, knowledge extraction, database reading, predicting material property, experiment design, computational scripts generation, guiding experiment, data analysis, and paper polishing, which covers the basic units of MOFs research. In general, these LLMs were capable of most of the tasks. Especially, Llama2-7B and ChatGLM2-6B were found to perform particularly well with moderate computational resources. Additionally, the performance of different parameter versions of the same model was compared, which revealed the superior performance of higher parameter versions.
Collapse
Affiliation(s)
- Xuefeng Bai
- Beijing Key Laboratory for Green Catalysis and Separation and Department of Chemical Engineering, College of Materials Science & Engineering, Beijing University of Technology, Beijing 100124, P. R. China
| | - Yabo Xie
- Beijing Key Laboratory for Green Catalysis and Separation and Department of Chemical Engineering, College of Materials Science & Engineering, Beijing University of Technology, Beijing 100124, P. R. China
| | - Xin Zhang
- Beijing Key Laboratory for Green Catalysis and Separation and Department of Chemical Engineering, College of Materials Science & Engineering, Beijing University of Technology, Beijing 100124, P. R. China
| | - Honggui Han
- Engineering Research Center of Digital Community, Ministry of Education, Beijing Laboratory for Urban Mass Transit and Beijing Key Laboratory of Computational Intelligence and Intelligence System, Faculty of Information Technology, Beijing University of Technology, Beijing 100124, P. R. China
| | - Jian-Rong Li
- Beijing Key Laboratory for Green Catalysis and Separation and Department of Chemical Engineering, College of Materials Science & Engineering, Beijing University of Technology, Beijing 100124, P. R. China
| |
Collapse
|
42
|
Li B, Raji IO, Gordon AGR, Sun L, Raimondo TM, Oladimeji FA, Jiang AY, Varley A, Langer RS, Anderson DG. Accelerating ionizable lipid discovery for mRNA delivery using machine learning and combinatorial chemistry. NATURE MATERIALS 2024; 23:1002-1008. [PMID: 38740955 DOI: 10.1038/s41563-024-01867-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 03/15/2024] [Indexed: 05/16/2024]
Abstract
To unlock the full promise of messenger (mRNA) therapies, expanding the toolkit of lipid nanoparticles is paramount. However, a pivotal component of lipid nanoparticle development that remains a bottleneck is identifying new ionizable lipids. Here we describe an accelerated approach to discovering effective ionizable lipids for mRNA delivery that combines machine learning with advanced combinatorial chemistry tools. Starting from a simple four-component reaction platform, we create a chemically diverse library of 584 ionizable lipids. We screen the mRNA transfection potencies of lipid nanoparticles containing those lipids and use the data as a foundational dataset for training various machine learning models. We choose the best-performing model to probe an expansive virtual library of 40,000 lipids, synthesizing and experimentally evaluating the top 16 lipids flagged. We identify lipid 119-23, which outperforms established benchmark lipids in transfecting muscle and immune cells in several tissues. This approach facilitates the creation and evaluation of versatile ionizable lipid libraries, advancing the formulation of lipid nanoparticles for precise mRNA delivery.
Collapse
Affiliation(s)
- Bowen Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada.
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada.
| | - Idris O Raji
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesiology, Boston Children's Hospital, Boston, MA, USA
| | - Akiva G R Gordon
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lizhuang Sun
- Department of Statistics, University of Michigan, Ann Arbor, MI, USA
| | - Theresa M Raimondo
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Favour A Oladimeji
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Allen Y Jiang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andrew Varley
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Robert S Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesiology, Boston Children's Hospital, Boston, MA, USA
- Department of Statistics, University of Michigan, Ann Arbor, MI, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Anesthesiology, Boston Children's Hospital, Boston, MA, USA.
- Department of Statistics, University of Michigan, Ann Arbor, MI, USA.
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
43
|
Ahmad S, Raza K. An extensive review on lung cancer therapeutics using machine learning techniques: state-of-the-art and perspectives. J Drug Target 2024; 32:635-646. [PMID: 38662768 DOI: 10.1080/1061186x.2024.2347358] [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] [Accepted: 04/18/2024] [Indexed: 05/07/2024]
Abstract
There are over 100 types of human cancer, accounting for millions of deaths every year. Lung cancer alone claims over 1.8 million lives per year and is expected to surpass 3.2 million by 2050, which underscores the urgent need for rapid drug development and repurposing initiatives. The application of AI emerges as a pivotal solution to developing anti-cancer therapeutics. This state-of-the-art review aims to explore the various applications of AI in lung cancer therapeutics. Predictive models can analyse large datasets, including clinical data, genetic information, and treatment outcomes, for novel drug design and to generate personalised treatment recommendations, potentially optimising therapeutic strategies, enhancing treatment efficacy, and minimising adverse effects. A thorough literature review study was conducted based on articles indexed in PubMed and Scopus. We compiled the use of various machine learning approaches, including CNN, RNN, GAN, VAEs, and other AI techniques, enhancing efficiency with accuracy exceeding 95%, which is validated through a computer-aided drug design process. AI can revolutionise lung cancer therapeutics, streamlining processes and saving biological scientists' time and effort-however, further research is needed to overcome challenges and fully unlock AI's potential in Lung Cancer Therapeutics.
Collapse
Affiliation(s)
- Shaban Ahmad
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Khalid Raza
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
44
|
Hao Y, Wang H, Liu X, Gai W, Hu S, Liu W, Miao Z, Gan Y, Yu X, Shi R, Tan Y, Kang T, Hai A, Zhao Y, Fu Y, Tang Y, Ye L, Liu J, Liang X, Ke B. Deep simulated annealing for the discovery of novel dental anesthetics with local anesthesia and anti-inflammatory properties. Acta Pharm Sin B 2024; 14:3086-3109. [PMID: 39027234 PMCID: PMC11252475 DOI: 10.1016/j.apsb.2024.01.019] [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: 11/22/2023] [Revised: 01/04/2024] [Accepted: 01/22/2024] [Indexed: 07/20/2024] Open
Abstract
Multifunctional therapeutics have emerged as a solution to the constraints imposed by drugs with singular or insufficient therapeutic effects. The primary challenge is to integrate diverse pharmacophores within a single-molecule framework. To address this, we introduced DeepSA, a novel edit-based generative framework that utilizes deep simulated annealing for the modification of articaine, a well-known local anesthetic. DeepSA integrates deep neural networks into metaheuristics, effectively constraining molecular space during compound generation. This framework employs a sophisticated objective function that accounts for scaffold preservation, anti-inflammatory properties, and covalent constraints. Through a sequence of local editing to navigate the molecular space, DeepSA successfully identified AT-17, a derivative exhibiting potent analgesic properties and significant anti-inflammatory activity in various animal models. Mechanistic insights into AT-17 revealed its dual mode of action: selective inhibition of NaV1.7 and 1.8 channels, contributing to its prolonged local anesthetic effects, and suppression of inflammatory mediators via modulation of the NLRP3 inflammasome pathway. These findings not only highlight the efficacy of AT-17 as a multifunctional drug candidate but also highlight the potential of DeepSA in facilitating AI-enhanced drug discovery, particularly within stringent chemical constraints.
Collapse
Affiliation(s)
- Yihang Hao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Haofan Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xianggen Liu
- College of Computer Science, Sichuan University, Chengdu 610065, China
| | - Wenrui Gai
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shilong Hu
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wencheng Liu
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhuang Miao
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yu Gan
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xianghua Yu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Rongjia Shi
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yongzhen Tan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ting Kang
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ao Hai
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi Zhao
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yihang Fu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yaling Tang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jin Liu
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinhua Liang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Bowen Ke
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
45
|
Snyder SH, Vignaux PA, Ozalp MK, Gerlach J, Puhl AC, Lane TR, Corbett J, Urbina F, Ekins S. The Goldilocks paradigm: comparing classical machine learning, large language models, and few-shot learning for drug discovery applications. Commun Chem 2024; 7:134. [PMID: 38866916 PMCID: PMC11169557 DOI: 10.1038/s42004-024-01220-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/04/2024] [Indexed: 06/14/2024] Open
Abstract
Recent advances in machine learning (ML) have led to newer model architectures including transformers (large language models, LLMs) showing state of the art results in text generation and image analysis as well as few-shot learning (FSLC) models which offer predictive power with extremely small datasets. These new architectures may offer promise, yet the 'no-free lunch' theorem suggests that no single model algorithm can outperform at all possible tasks. Here, we explore the capabilities of classical (SVR), FSLC, and transformer models (MolBART) over a range of dataset tasks and show a 'goldilocks zone' for each model type, in which dataset size and feature distribution (i.e. dataset "diversity") determines the optimal algorithm strategy. When datasets are small ( < 50 molecules), FSLC tend to outperform both classical ML and transformers. When datasets are small-to-medium sized (50-240 molecules) and diverse, transformers outperform both classical models and few-shot learning. Finally, when datasets are of larger and of sufficient size, classical models then perform the best, suggesting that the optimal model to choose likely depends on the dataset available, its size and diversity. These findings may help to answer the perennial question of which ML algorithm is to be used when faced with a new dataset.
Collapse
Affiliation(s)
- Scott H Snyder
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Patricia A Vignaux
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Mustafa Kemal Ozalp
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Jacob Gerlach
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Ana C Puhl
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Thomas R Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - John Corbett
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA
| | - Fabio Urbina
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA.
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, USA.
| |
Collapse
|
46
|
Zhang D, Zhao D, Wang Z, Li J, Li J. MMSSC-Net: multi-stage sequence cognitive networks for drug molecule recognition. RSC Adv 2024; 14:18182-18191. [PMID: 38854833 PMCID: PMC11155551 DOI: 10.1039/d4ra02442g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 04/29/2024] [Indexed: 06/11/2024] Open
Abstract
In the growing body of scientific literature, the structure and information of drugs are usually represented in two-dimensional vector graphics. Drug compound structures in vector graphics form are difficult to recognize and utilize by computers. Although the current OCSR paradigm has shown good performance, most existing work treats it as a single isolated whole. This paper proposes a multi-stage cognitive neural network model that predicts molecular vector graphics more finely. Based on cognitive methods, we construct a model for fine-grained perceptual representation of molecular images from bottom to top, and in stages, the primary representation of atoms and bonds is potential discrete label sequence (atom type, bond type, functional group, etc.). The second stage represents the molecular graph according to the label sequence, and the final stage evolves in an extensible manner from the molecular graph to a machine-readable sequence. Experimental results show that MMSSC-Net outperforms current advanced methods on multiple public datasets. It achieved an accuracy rate of 75-94% on cognitive recognition at different resolutions. MMSSC-Net uses a sequence cognitive method to make it more reliable in interpretability and transferability, and provides new ideas for drug information discovery and exploring the unknown chemical space.
Collapse
Affiliation(s)
- Dehai Zhang
- The Key Laboratory of Software Engineering of Yunnan Province, School of Software, Yunnan University Kunming China
| | - Di Zhao
- The Key Laboratory of Software Engineering of Yunnan Province, School of Software, Yunnan University Kunming China
| | - Zhengwu Wang
- The Key Laboratory of Software Engineering of Yunnan Province, School of Software, Yunnan University Kunming China
| | - Junhui Li
- The Key Laboratory of Software Engineering of Yunnan Province, School of Software, Yunnan University Kunming China
| | - Jin Li
- The Key Laboratory of Software Engineering of Yunnan Province, School of Software, Yunnan University Kunming China
| |
Collapse
|
47
|
Tian T, Li S, Zhang Z, Chen L, Zou Z, Zhao D, Zeng J. Benchmarking compound activity prediction for real-world drug discovery applications. Commun Chem 2024; 7:127. [PMID: 38834746 DOI: 10.1038/s42004-024-01204-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/16/2024] [Indexed: 06/06/2024] Open
Abstract
Identifying active compounds for target proteins is fundamental in early drug discovery. Recently, data-driven computational methods have demonstrated promising potential in predicting compound activities. However, there lacks a well-designed benchmark to comprehensively evaluate these methods from a practical perspective. To fill this gap, we propose a Compound Activity benchmark for Real-world Applications (CARA). Through carefully distinguishing assay types, designing train-test splitting schemes and selecting evaluation metrics, CARA can consider the biased distribution of current real-world compound activity data and avoid overestimation of model performances. We observed that although current models can make successful predictions for certain proportions of assays, their performances varied across different assays. In addition, evaluation of several few-shot training strategies demonstrated different performances related to task types. Overall, we provide a high-quality dataset for developing and evaluating compound activity prediction models, and the analyses in this work may inspire better applications of data-driven models in drug discovery.
Collapse
Affiliation(s)
- Tingzhong Tian
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing, China
| | - Shuya Li
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing, China
| | - Ziting Zhang
- Department of Automation, Tsinghua University, Beijing, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing, China
| | - Lin Chen
- Silexon AI Technology Co., Ltd., Nanjing, Jiangsu Province, China
| | - Ziheng Zou
- Silexon AI Technology Co., Ltd., Nanjing, Jiangsu Province, China
| | - Dan Zhao
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing, China.
| | - Jianyang Zeng
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing, China.
- School of Engineering, Westlake University, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
48
|
Balog S, de Almeida MS, Taladriz-Blanco P, Rothen-Rutishauser B, Petri-Fink A. Does the surface charge of the nanoparticles drive nanoparticle-cell membrane interactions? Curr Opin Biotechnol 2024; 87:103128. [PMID: 38581743 DOI: 10.1016/j.copbio.2024.103128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/08/2024] [Accepted: 03/13/2024] [Indexed: 04/08/2024]
Abstract
Classical Coulombic interaction, characterized by electrostatic interactions mediated through surface charges, is often regarded as the primary determinant in nanoparticles' (NPs) cellular association and internalization. However, the intricate physicochemical properties of particle surfaces, biomolecular coronas, and cell surfaces defy this oversimplified perspective. Moreover, the nanometrological techniques employed to characterize NPs in complex physiological fluids often exhibit limited accuracy and reproducibility. A more comprehensive understanding of nanoparticle-cell membrane interactions, extending beyond attractive forces between oppositely charged surfaces, necessitates the establishment of databases through rigorous physical, chemical, and biological characterization supported by nanoscale analytics. Additionally, computational approaches, such as in silico modeling and machine learning, play a crucial role in unraveling the complexities of these interactions.
Collapse
Affiliation(s)
- Sandor Balog
- Adolphe Merkle Institute, University of Fribourg, National Center of Competence in Research Bio-Inspired Materials, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Mauro Sousa de Almeida
- Adolphe Merkle Institute, University of Fribourg, National Center of Competence in Research Bio-Inspired Materials, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Patricia Taladriz-Blanco
- Adolphe Merkle Institute, University of Fribourg, National Center of Competence in Research Bio-Inspired Materials, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Barbara Rothen-Rutishauser
- Adolphe Merkle Institute, University of Fribourg, National Center of Competence in Research Bio-Inspired Materials, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Alke Petri-Fink
- Adolphe Merkle Institute, University of Fribourg, National Center of Competence in Research Bio-Inspired Materials, Chemin des Verdiers 4, 1700 Fribourg, Switzerland; Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland.
| |
Collapse
|
49
|
Chen T, Pang Z, He S, Li Y, Shrestha S, Little JM, Yang H, Chung TC, Sun J, Whitley HC, Lee IC, Woehl TJ, Li T, Hu L, Chen PY. Machine intelligence-accelerated discovery of all-natural plastic substitutes. NATURE NANOTECHNOLOGY 2024; 19:782-791. [PMID: 38499859 PMCID: PMC11186784 DOI: 10.1038/s41565-024-01635-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 02/15/2024] [Indexed: 03/20/2024]
Abstract
One possible solution against the accumulation of petrochemical plastics in natural environments is to develop biodegradable plastic substitutes using natural components. However, discovering all-natural alternatives that meet specific properties, such as optical transparency, fire retardancy and mechanical resilience, which have made petrochemical plastics successful, remains challenging. Current approaches still rely on iterative optimization experiments. Here we show an integrated workflow that combines robotics and machine learning to accelerate the discovery of all-natural plastic substitutes with programmable optical, thermal and mechanical properties. First, an automated pipetting robot is commanded to prepare 286 nanocomposite films with various properties to train a support-vector machine classifier. Next, through 14 active learning loops with data augmentation, 135 all-natural nanocomposites are fabricated stagewise, establishing an artificial neural network prediction model. We demonstrate that the prediction model can conduct a two-way design task: (1) predicting the physicochemical properties of an all-natural nanocomposite from its composition and (2) automating the inverse design of biodegradable plastic substitutes that fulfils various user-specific requirements. By harnessing the model's prediction capabilities, we prepare several all-natural substitutes, that could replace non-biodegradable counterparts as exhibiting analogous properties. Our methodology integrates robot-assisted experiments, machine intelligence and simulation tools to accelerate the discovery and design of eco-friendly plastic substitutes starting from building blocks taken from the generally-recognized-as-safe database.
Collapse
Affiliation(s)
- Tianle Chen
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD, USA
| | - Zhenqian Pang
- Department of Mechanical Engineering, University of Maryland, College Park, MD, USA
| | - Shuaiming He
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD, USA
| | - Yang Li
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD, USA
| | - Snehi Shrestha
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD, USA
| | - Joshua M Little
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD, USA
| | - Haochen Yang
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD, USA
| | - Tsai-Chun Chung
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD, USA
| | - Jiayue Sun
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, USA
| | | | - I-Chi Lee
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Taylor J Woehl
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD, USA
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, USA
| | - Teng Li
- Department of Mechanical Engineering, University of Maryland, College Park, MD, USA.
| | - Liangbing Hu
- Department of Materials Science and Engineering, University of Maryland, College Park, MD, USA.
| | - Po-Yen Chen
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD, USA.
- Maryland Robotics Center, College Park, MD, USA.
| |
Collapse
|
50
|
Taub R, Savir Y. SAF: Smart Aggregation Framework for Revealing Atoms Importance Rank and Improving Prediction Rates in Drug Discovery. J Chem Inf Model 2024; 64:4021-4030. [PMID: 38695490 PMCID: PMC11134513 DOI: 10.1021/acs.jcim.4c00107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 05/28/2024]
Abstract
Machine learning, and representation learning in particular, has the potential to facilitate drug discovery by screening a large chemical space in silico. A successful approach for representing molecules is to treat them as graphs and utilize graph neural networks. One of the key limitations of such methods is the necessity to represent compounds with different numbers of atoms, which requires aggregating the atom's information. Common aggregation operators, such as averaging, result in a loss of information at the atom level. In this work, we propose a novel aggregating approach where each atom is weighted nonlinearly using the Boltzmann distribution with a hyperparameter analogous to temperature. We show that using this weighted aggregation improves the ability of the gold standard message-passing neural network to predict antibiotic activity. Moreover, by changing the temperature hyperparameter, our approach can reveal the atoms that are important for activity prediction in a smooth and consistent way, thus providing a novel regulated attention mechanism for graph neural networks. We further validate our method by showing that it recapitulates the functional group in β-lactam antibiotics. The ability of our approach to rank the atoms' importance for a desired function can be used within any graph neural network to provide interpretability of the results and predictions at the node level.
Collapse
Affiliation(s)
- Ronen Taub
- Department of Physiology, Biophysics
& Systems Biology, Medicine Faculty, Technion IIT, Haifa 3525422, Israel
| | - Yonatan Savir
- Department of Physiology, Biophysics
& Systems Biology, Medicine Faculty, Technion IIT, Haifa 3525422, Israel
| |
Collapse
|