1
|
Cobre ADF, Fachi MM, Domingues KZA, Lazo REL, Ferreira LM, Tonin FS, Pontarolo R. Accuracy of COVID-19 diagnostic tests via infrared spectroscopy: A systematic review and meta-analysis. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 327:125337. [PMID: 39481165 DOI: 10.1016/j.saa.2024.125337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024]
Abstract
This study aims to synthesize the evidence on the accuracy parameters of COVID-19 diagnosis methods using infrared spectroscopy (FTIR). A systematic review with searches in PubMed and Embase was performed (September 2023). Studies reporting data on test specificity, sensitivity, true positive, true negative, false positive, and false negative using different human samples were included. Meta-analysis of accuracy estimates with 95 % confidence intervals and area under the ROC Curve (AUC) were conducted (Meta-Disc 1.4.7). Seventeen studies were included - all of them highlighted regions 650-1800 cm-1 and 2300-3900 cm-1 as most important for diagnosing COVID-19. The FTIR technique presented high sensitivity [0.912 (95 %CI, 0.878-0.939), especially in vaccinated [0.959 (CI95 %, 0.908-0.987)] compared to unvaccinated [0.625 (CI95 %, 0.584-0.664)] individuals for COVID-19. Overall specificity was also high [0.886 (95 %CI, 0.855-0.912), with increased rates in vaccinated [0.884 (CI95 %, 0.819-0.932)] than in unvaccinated [0.667 (CI95 %, 0.629-0.704)] patients. These findings reveal that FTIR is an accurate technique for detecting SARS-CoV-2 infection in different biological matrices with advantages including low cost, rapid and environmentally friendly with minimal preparation analyses. This could lead to an easy implementation of this technique in practice as a screening tool for patients with suspected COVID-19, especially in low-income countries.
Collapse
Affiliation(s)
- Alexandre de Fátima Cobre
- Pharmaceutical Sciences Postgraduate Program, Universidade Federal do Paraná, Curitiba, Brazil; School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, United Kingdom
| | - Mariana Millan Fachi
- Pharmaceutical Sciences Postgraduate Program, Universidade Federal do Paraná, Curitiba, Brazil
| | | | - Raul Edison Luna Lazo
- Pharmaceutical Sciences Postgraduate Program, Universidade Federal do Paraná, Curitiba, Brazil
| | - Luana Mota Ferreira
- Department of Pharmacy, Pharmaceutical Sciences Postgraduate Program, Universidade Federal do Paraná, Curitiba, Brazil
| | - Fernanda Stumpf Tonin
- H&TRC - Health & Technology Research Centre, ESTeSL, Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Lisbon, Portugal
| | - Roberto Pontarolo
- Department of Pharmacy, Pharmaceutical Sciences Postgraduate Program, Universidade Federal do Paraná, Curitiba, Brazil.
| |
Collapse
|
2
|
Serrano DR, Luciano FC, Anaya BJ, Ongoren B, Kara A, Molina G, Ramirez BI, Sánchez-Guirales SA, Simon JA, Tomietto G, Rapti C, Ruiz HK, Rawat S, Kumar D, Lalatsa A. Artificial Intelligence (AI) Applications in Drug Discovery and Drug Delivery: Revolutionizing Personalized Medicine. Pharmaceutics 2024; 16:1328. [PMID: 39458657 PMCID: PMC11510778 DOI: 10.3390/pharmaceutics16101328] [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: 08/19/2024] [Revised: 10/06/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Artificial intelligence (AI) encompasses a broad spectrum of techniques that have been utilized by pharmaceutical companies for decades, including machine learning, deep learning, and other advanced computational methods. These innovations have unlocked unprecedented opportunities for the acceleration of drug discovery and delivery, the optimization of treatment regimens, and the improvement of patient outcomes. AI is swiftly transforming the pharmaceutical industry, revolutionizing everything from drug development and discovery to personalized medicine, including target identification and validation, selection of excipients, prediction of the synthetic route, supply chain optimization, monitoring during continuous manufacturing processes, or predictive maintenance, among others. While the integration of AI promises to enhance efficiency, reduce costs, and improve both medicines and patient health, it also raises important questions from a regulatory point of view. In this review article, we will present a comprehensive overview of AI's applications in the pharmaceutical industry, covering areas such as drug discovery, target optimization, personalized medicine, drug safety, and more. By analyzing current research trends and case studies, we aim to shed light on AI's transformative impact on the pharmaceutical industry and its broader implications for healthcare.
Collapse
Affiliation(s)
- Dolores R. Serrano
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
- Instituto Universitario de Farmacia Industrial, 28040 Madrid, Spain
| | - Francis C. Luciano
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Brayan J. Anaya
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Baris Ongoren
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Aytug Kara
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Gracia Molina
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Bianca I. Ramirez
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Sergio A. Sánchez-Guirales
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Jesus A. Simon
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Greta Tomietto
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Chrysi Rapti
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Helga K. Ruiz
- Department of Pharmaceutics and Food Science, School of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (F.C.L.); (B.J.A.); (B.O.); (A.K.); (G.M.); (B.I.R.); (S.A.S.-G.); (J.A.S.); (G.T.); (C.R.); (H.K.R.)
| | - Satyavati Rawat
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, India; (S.R.); (D.K.)
| | - Dinesh Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, India; (S.R.); (D.K.)
| | - Aikaterini Lalatsa
- Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161, Cathedral Street, Glasgow G4 0RE, UK
- CRUK Formulation Unit, School of Pharmacy and Biomedical Sciences, University of Strathclyde, 161, Cathedral Street, Glasgow G4 0RE, UK
| |
Collapse
|
3
|
Munir N, de Lima T, Nugent M, McAfee M. In-line NIR coupled with machine learning to predict mechanical properties and dissolution profile of PLA-Aspirin. FUNCTIONAL COMPOSITE MATERIALS 2024; 5:14. [PMID: 39391170 PMCID: PMC11461551 DOI: 10.1186/s42252-024-00063-5] [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: 06/01/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024]
Abstract
In the production of polymeric drug delivery devices, dissolution profile and mechanical properties of the drug loaded polymeric matrix are considered important Critical Quality Attributes (CQA) for quality assurance. However, currently the industry relies on offline testing methods which are destructive, slow, labour intensive, and costly. In this work, a real-time method for predicting these CQAs in a Hot Melt Extrusion (HME) process is explored using in-line NIR and temperature sensors together with Machine Learning (ML) algorithms. The mechanical and drug dissolution properties were found to vary significantly with changes in processing conditions, highlighting that real-time methods to accurately predict product properties are highly desirable for process monitoring and optimisation. Nonlinear ML methods including Random Forest (RF), K-Nearest Neighbours (KNN) and Recursive Feature Elimination with RF (RFE-RF) outperformed commonly used linear machine learning methods. For the prediction of tensile strength RFE-RF and KNN achieved R 2 values 98% and 99%, respectively. For the prediction of drug dissolution, two time points were considered with drug release at t = 6 h as a measure of the extent of burst release, and t = 96 h as a measure of sustained release. KNN and RFE-RF achieved R 2 values of 97% and 96%, respectively in predicting the drug release at t = 96 h. This work for the first time reports the prediction of drug dissolution and mechanical properties of drug loaded polymer product from in-line data collected during the HME process. Supplementary Information The online version contains supplementary material available at 10.1186/s42252-024-00063-5.
Collapse
Affiliation(s)
- Nimra Munir
- Centre for Mathematical Modelling and Intelligent Systems for Health and Environment (MISHE), Atlantic Technological University, ATU Sligo, Ash Lane, Co. Sligo F91 YW50 Ireland
- Centre for Precision Engineering, Materials and Manufacturing (PEM Centre), Atlantic Technological University, ATU Sligo, Ash Lane, Co. Sligo F91 YW50 Ireland
| | - Tielidy de Lima
- Materials Research Institute, Technological University of the Shannon: Midlands Midwest, Athlone, N37HD68 Ireland
| | - Michael Nugent
- Materials Research Institute, Technological University of the Shannon: Midlands Midwest, Athlone, N37HD68 Ireland
| | - Marion McAfee
- Centre for Mathematical Modelling and Intelligent Systems for Health and Environment (MISHE), Atlantic Technological University, ATU Sligo, Ash Lane, Co. Sligo F91 YW50 Ireland
- Centre for Precision Engineering, Materials and Manufacturing (PEM Centre), Atlantic Technological University, ATU Sligo, Ash Lane, Co. Sligo F91 YW50 Ireland
| |
Collapse
|
4
|
Suriyaamporn P, Pamornpathomkul B, Patrojanasophon P, Ngawhirunpat T, Rojanarata T, Opanasopit P. The Artificial Intelligence-Powered New Era in Pharmaceutical Research and Development: A Review. AAPS PharmSciTech 2024; 25:188. [PMID: 39147952 DOI: 10.1208/s12249-024-02901-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 07/22/2024] [Indexed: 08/17/2024] Open
Abstract
Currently, artificial intelligence (AI), machine learning (ML), and deep learning (DL) are gaining increased interest in many fields, particularly in pharmaceutical research and development, where they assist in decision-making in complex situations. Numerous research studies and advancements have demonstrated how these computational technologies are used in various pharmaceutical research and development aspects, including drug discovery, personalized medicine, drug formulation, optimization, predictions, drug interactions, pharmacokinetics/ pharmacodynamics, quality control/quality assurance, and manufacturing processes. Using advanced modeling techniques, these computational technologies can enhance efficiency and accuracy, handle complex data, and facilitate novel discoveries within minutes. Furthermore, these technologies offer several advantages over conventional statistics. They allow for pattern recognition from complex datasets, and the models, typically developed from data-driven algorithms, can predict a given outcome (model output) from a set of features (model inputs). Additionally, this review discusses emerging trends and provides perspectives on the application of AI with quality by design (QbD) and the future role of AI in this field. Ethical and regulatory considerations associated with integrating AI into pharmaceutical technology were also examined. This review aims to offer insights to researchers, professionals, and others on the current state of AI applications in pharmaceutical research and development and their potential role in the future of research and the era of pharmaceutical Industry 4.0 and 5.0.
Collapse
Affiliation(s)
- Phuvamin Suriyaamporn
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Boonnada Pamornpathomkul
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Prasopchai Patrojanasophon
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Tanasait Ngawhirunpat
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Theerasak Rojanarata
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Praneet Opanasopit
- Pharmaceutical Development of Green Innovations Group (PDGIG), Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand.
| |
Collapse
|
5
|
Gholap AD, Uddin MJ, Faiyazuddin M, Omri A, Gowri S, Khalid M. Advances in artificial intelligence for drug delivery and development: A comprehensive review. Comput Biol Med 2024; 178:108702. [PMID: 38878397 DOI: 10.1016/j.compbiomed.2024.108702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/12/2024] [Accepted: 06/01/2024] [Indexed: 07/24/2024]
Abstract
Artificial intelligence (AI) has emerged as a powerful tool to revolutionize the healthcare sector, including drug delivery and development. This review explores the current and future applications of AI in the pharmaceutical industry, focusing on drug delivery and development. It covers various aspects such as smart drug delivery networks, sensors, drug repurposing, statistical modeling, and simulation of biotechnological and biological systems. The integration of AI with nanotechnologies and nanomedicines is also examined. AI offers significant advancements in drug discovery by efficiently identifying compounds, validating drug targets, streamlining drug structures, and prioritizing response templates. Techniques like data mining, multitask learning, and high-throughput screening contribute to better drug discovery and development innovations. The review discusses AI applications in drug formulation and delivery, clinical trials, drug safety, and pharmacovigilance. It addresses regulatory considerations and challenges associated with AI in pharmaceuticals, including privacy, data security, and interpretability of AI models. The review concludes with future perspectives, highlighting emerging trends, addressing limitations and biases in AI models, and emphasizing the importance of collaboration and knowledge sharing. It provides a comprehensive overview of AI's potential to transform the pharmaceutical industry and improve patient care while identifying further research and development areas.
Collapse
Affiliation(s)
- Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar, Maharashtra, 401404, India.
| | - Md Jasim Uddin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Md Faiyazuddin
- School of Pharmacy, Al-Karim University, Katihar, Bihar, 854106, India; Centre for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Tamil Nadu, India.
| | - Abdelwahab Omri
- Department of Chemistry and Biochemistry, The Novel Drug and Vaccine Delivery Systems Facility, Laurentian University, Sudbury, ON, P3E 2C6, Canada.
| | - S Gowri
- PG & Research, Department of Physics, Cauvery College for Women, Tiruchirapalli, Tamil Nadu, 620018, India
| | - Mohammad Khalid
- James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK; Sunway Centre for Electrochemical Energy and Sustainable Technology (SCEEST), School of Engineering and Technology, Sunway University, No. 5, Jalan Universiti, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia; University Centre for Research and Development, Chandigarh University, Mohali, Punjab, 140413, India.
| |
Collapse
|
6
|
Wang L, Zhou Z, Yang X, Shi S, Zeng X, Cao D. The present state and challenges of active learning in drug discovery. Drug Discov Today 2024; 29:103985. [PMID: 38642700 DOI: 10.1016/j.drudis.2024.103985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 04/22/2024]
Abstract
Active learning (AL) is an iterative feedback process that efficiently identifies valuable data within vast chemical space, even with limited labeled data. This characteristic renders it a valuable approach to tackle the ongoing challenges faced in drug discovery, such as the ever-expanding explore space and the limitations of labeled data. Consequently, AL is increasingly gaining prominence in the field of drug development. In this paper, we comprehensively review the application of AL at all stages of drug discovery, including compounds-target interaction prediction, virtual screening, molecular generation and optimization, as well as molecular properties prediction. Additionally, we discuss the challenges and prospects associated with the current applications of AL in drug discovery.
Collapse
Affiliation(s)
- Lei Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Zhenran Zhou
- Department of Computer Science, Hunan University, Changsha 410082, Hunan, China
| | - Xixi Yang
- Department of Computer Science, Hunan University, Changsha 410082, Hunan, China
| | - Shaohua Shi
- Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xiangxiang Zeng
- Department of Computer Science, Hunan University, Changsha 410082, Hunan, China.
| | - Dongsheng Cao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China.
| |
Collapse
|
7
|
Yang M, Meng Y, Hao W, Zhang J, Liu J, Wu L, Lin B, Liu Y, Zhang Y, Yu X, Wang X, Gong Y, Ge L, Fan Y, Xie C, Xu Y, Chang Q, Zhang Y, Qin X. A prognostic model for SARS-CoV-2 breakthrough infection: Analyzing a prospective cellular immunity cohort. Int Immunopharmacol 2024; 131:111829. [PMID: 38489974 DOI: 10.1016/j.intimp.2024.111829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/03/2024] [Accepted: 03/06/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND Following the COVID-19 pandemic, studies have identified several prevalent characteristics, especially related to lymphocyte subsets. However, limited research is available on the focus of this study, namely, the specific memory cell subsets among individuals who received COVID-19 vaccine boosters and subsequently experienced a SARS-CoV-2 breakthrough infection. METHODS Flow cytometry (FCM) was employed to investigate the early and longitudinal pattern changes of cellular immunity in patients with SARS-CoV-2 breakthrough infections following COVID-19 vaccine boosters. XGBoost (a machine learning algorithm) was employed to analyze cellular immunity prior to SARS-CoV-2 breakthrough, aiming to establish a prognostic model for SARS-CoV-2 breakthrough infections. RESULTS Following SARS-CoV-2 breakthrough infection, naïve T cells and TEMRA subsets increased while the percentage of TCM and TEM cells decreased. Naïve and non-switched memory B cells increased while switched and double-negative memory B cells decreased. The XGBoost model achieved an area under the curve (AUC) of 0.78, with an accuracy rate of 81.8 %, a sensitivity of 75 %, and specificity of 85.7 %. TNF-α, CD27-CD19+cells, and TEMRA subsets were identified as high predictors. An increase in TNF-α, cTfh, double-negative memory B cells, IL-6, IL-10, and IFN-γ prior to SARS-CoV-2 infection was associated with enduring clinical symptoms; conversely, an increase in CD3+ T cells, CD4+ T cells, and IL-2 was associated with clinical with non-enduring clinical symptoms. CONCLUSION SARS-CoV-2 breakthrough infection leads to disturbances in cellular immunity. Assessing cellular immunity prior to breakthrough infection serves as a valuable prognostic tool for SARS-CoV-2 infection, which facilitates clinical decision-making.
Collapse
Affiliation(s)
- Mei Yang
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yuan Meng
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Wudi Hao
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Jin Zhang
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Jianhua Liu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Lina Wu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Baoxu Lin
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yong Liu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yue Zhang
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiaojun Yu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiaoqian Wang
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yu Gong
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Lili Ge
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yan Fan
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Conghong Xie
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yiyun Xu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Qing Chang
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yixiao Zhang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| | - Xiaosong Qin
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
8
|
Yang R, Fu Y, Zhang Q, Zhang L. GCNGAT: Drug-disease association prediction based on graph convolution neural network and graph attention network. Artif Intell Med 2024; 150:102805. [PMID: 38553169 DOI: 10.1016/j.artmed.2024.102805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 01/22/2024] [Accepted: 02/08/2024] [Indexed: 04/02/2024]
Abstract
Predicting drug-disease associations can contribute to discovering new therapeutic potentials of drugs, and providing important association information for new drug research and development. Many existing drug-disease association prediction methods have not distinguished relevant background information for the same drug targeted to different diseases. Therefore, this paper proposes a drug-disease association prediction model based on graph convolutional network and graph attention network (GCNGAT) to reposition marketed drugs under the distinguishment of background information. Firstly, in order to obtain initial drug-disease information, a drug-disease heterogeneous graph structure is constructed based on all known drug-disease associations. Secondly, based on the heterogeneous graph structure, the corresponding subgraphs of each group of drug-disease association pairs are extracted to distinguish different background information for the same drug from different diseases. Finally, a model combining Graph neural network with global Average pooling (GnnAp) is designed to predict potential drug-disease associations by learning drug-disease interaction feature representations. The experimental results show that adding subgraph extraction can effectively improve the prediction performance of the model, and the graph representation learning module can fully extract the deep features of drug-disease. Using the 5-fold cross-validation, the proposed model (GCNGAT) achieves AUC (Area Under the receiver operating characteristic Curve) values of 0.9182 and 0.9417 on the PREDICT dataset and CDataset dataset, respectively. Compared with other predictors on the same dataset (PREDICT dataset), GCNGAT outperforms the existing best-performing model (PSGCN), with a 1.58% increase in the AUC value. It is anticipated that this model can provide experimental reference for drug repositioning and further promote the drug research and development process.
Collapse
Affiliation(s)
- Runtao Yang
- School of Mechanical, Electrical and Information Engineering, Shandong University at Weihai, 264209, China.
| | - Yao Fu
- School of Mechanical, Electrical and Information Engineering, Shandong University at Weihai, 264209, China.
| | - Qian Zhang
- Heze Institute of Science and Technology Information, Heze, 274000, China.
| | - Lina Zhang
- School of Mechanical, Electrical and Information Engineering, Shandong University at Weihai, 264209, China.
| |
Collapse
|
9
|
Sahriar S, Akther S, Mauya J, Amin R, Mia MS, Ruhi S, Reza MS. Unlocking stroke prediction: Harnessing projection-based statistical feature extraction with ML algorithms. Heliyon 2024; 10:e27411. [PMID: 38495193 PMCID: PMC10943390 DOI: 10.1016/j.heliyon.2024.e27411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/19/2024] Open
Abstract
Non-communicable diseases, such as cardiovascular disease, cancer, chronic respiratory diseases, and diabetes, are responsible for approximately 71% of all deaths worldwide. Stroke, a cerebrovascular disorder, is one of the leading contributors to this burden among the top three causes of death. Early recognition of symptoms can encourage a balanced lifestyle and provide essential information for stroke prediction. To identify a stroke patient and risk factors, machine learning (ML) is a key tool for physicians. Due to different data measurement scales and their probability distributional assumptions, ML-based algorithms struggle to detect risk factors. Furthermore, when dealing with risk factors with high-dimensional features, learning algorithms struggle with complexity. In this study, rigorous statistical tests are used to identify risk factors, and PCA-FA (Integration of Principal Components and Factors) and FPCA (Factor Based PCA) approaches are proposed for projecting suitable feature representations for improving learning algorithm performances. The study dataset consists of different clinical, lifestyle, and genetic attributes, allowing for a comprehensive analysis of potential risk factors associated with stroke, which contains 5110 patient records. Using significant test (P-value <0.05), chi-square and independent sample t-test identified age, heart_disease, hypertension, work_type, ever_married, bmi, and smoking_status as risk factors for stroke. To develop the predicting model with proposed feature extraction techniques, random forests approach provides the best results when utilizing the PCA-FA method. The best accuracy rate for this approach is 92.55%, while the AUC score is 98.15%. The prediction accuracy has increased from 2.19% to 19.03% compared to the existing work. Additionally, the prediction results is robustified and reproducible with a stacking ensemble-based classification algorithm. We also developed a web-based application to help doctors diagnose stroke risk based on the findings of this study, which could be used as an additional tool to help doctors diagnose.
Collapse
Affiliation(s)
- Saad Sahriar
- Deep Statistical Learning and Research Lab, Department of Statistics, Pabna University of Science & Technology, Pabna, 6600, Bangladesh
| | - Sanjida Akther
- Deep Statistical Learning and Research Lab, Department of Statistics, Pabna University of Science & Technology, Pabna, 6600, Bangladesh
| | - Jannatul Mauya
- Deep Statistical Learning and Research Lab, Department of Statistics, Pabna University of Science & Technology, Pabna, 6600, Bangladesh
| | - Ruhul Amin
- Deep Statistical Learning and Research Lab, Department of Statistics, Pabna University of Science & Technology, Pabna, 6600, Bangladesh
| | - Md Shahajada Mia
- Department of Statistics, Pabna University of Science & Technology, Pabna, 6600, Bangladesh
| | - Sabba Ruhi
- Department of Statistics, Pabna University of Science & Technology, Pabna, 6600, Bangladesh
| | - Md Shamim Reza
- Deep Statistical Learning and Research Lab, Department of Statistics, Pabna University of Science & Technology, Pabna, 6600, Bangladesh
- Department of Statistics, Pabna University of Science & Technology, Pabna, 6600, Bangladesh
| |
Collapse
|
10
|
Hadiby S, Ben Ali YM. Integrating pharmacophore model and deep learning for activity prediction of molecules with BRCA1 gene. J Bioinform Comput Biol 2024; 22:2450003. [PMID: 38567386 DOI: 10.1142/s0219720024500033] [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: 04/04/2024]
Abstract
In this paper, we propose a novel approach for predicting the activity/inactivity of molecules with the BRCA1 gene by combining pharmacophore modeling and deep learning techniques. Initially, we generated 3D pharmacophore fingerprints using a pharmacophore model, which captures the essential features and spatial arrangements critical for biological activity. These fingerprints served as informative representations of the molecular structures. Next, we employed deep learning algorithms to train a predictive model using the generated pharmacophore fingerprints. The deep learning model was designed to learn complex patterns and relationships between the pharmacophore features and the corresponding activity/inactivity labels of the molecules. By utilizing this integrated approach, we aimed to enhance the accuracy and efficiency of activity prediction. To validate the effectiveness of our approach, we conducted experiments using a dataset of known molecules with BRCA1 gene activity/inactivity from diverse sources. Our results demonstrated promising predictive performance, indicating the successful integration of pharmacophore modeling and deep learning. Furthermore, we utilized the trained model to predict the activity/inactivity of unknown molecules extracted from the ChEMBL database. The predictions obtained from the ChEMBL database were assessed and compared against experimentally determined values to evaluate the reliability and generalizability of our model. Overall, our proposed approach showcased significant potential in accurately predicting the activity/inactivity of molecules with the BRCA1 gene, thus enabling the identification of potential candidates for further investigation in drug discovery and development processes.
Collapse
Affiliation(s)
- Seloua Hadiby
- Department of Computer Science, Computer Research Laboratory, Badji Mokhtar University, Annaba, Algeria
| | - Yamina Mohamed Ben Ali
- Department of Computer Science, Computer Research Laboratory, Badji Mokhtar University, Annaba, Algeria
| |
Collapse
|
11
|
Srivastava S, Jain P. Computational Approaches: A New Frontier in Cancer Research. Comb Chem High Throughput Screen 2024; 27:1861-1876. [PMID: 38031782 DOI: 10.2174/0113862073265604231106112203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/08/2023] [Accepted: 09/21/2023] [Indexed: 12/01/2023]
Abstract
Cancer is a broad category of disease that can start in virtually any organ or tissue of the body when aberrant cells assault surrounding organs and proliferate uncontrollably. According to the most recent statistics, cancer will be the cause of 10 million deaths worldwide in 2020, accounting for one death out of every six worldwide. The typical approach used in anti-cancer research is highly time-consuming and expensive, and the outcomes are not particularly encouraging. Computational techniques have been employed in anti-cancer research to advance our understanding. Recent years have seen a significant and exceptional impact on anticancer research due to the rapid development of computational tools for novel drug discovery, drug design, genetic studies, genome characterization, cancer imaging and detection, radiotherapy, cancer metabolomics, and novel therapeutic approaches. In this paper, we examined the various subfields of contemporary computational techniques, including molecular docking, artificial intelligence, bioinformatics, virtual screening, and QSAR, and their applications in the study of cancer.
Collapse
Affiliation(s)
- Shubham Srivastava
- Department of Pharmacy, IIMT College of Pharmacy, Uttar Pradesh, 201310, India
| | - Pushpendra Jain
- Department of Pharmacy, IIMT College of Pharmacy, Uttar Pradesh, 201310, India
| |
Collapse
|
12
|
Hashizume T, Ying BW. Challenges in developing cell culture media using machine learning. Biotechnol Adv 2024; 70:108293. [PMID: 37984683 DOI: 10.1016/j.biotechadv.2023.108293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 10/17/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023]
Abstract
Microbial and mammalian cells are widely used in the food, pharmaceutical, and medical industries. Developing or optimizing culture media is essential to improve cell culture performance as a critical technology in cell culture engineering. Methodologies for media optimization have been developed to a great extent, such as the approaches of one-factor-at-a-time (OFAT) and response surface methodology (RSM). The present review introduces the emerging machine learning (ML) technology in cell culture engineering by combining high-throughput experimental technologies to develop highly efficient and effective culture media. The commonly used ML algorithms and the successful applications of employing ML in medium optimization are summarized. This review highlights the benefits of ML-assisted medium development and guides the selection of the media optimization method appropriate for various cell culture purposes.
Collapse
Affiliation(s)
- Takamasa Hashizume
- School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8572 Ibaraki, Japan
| | - Bei-Wen Ying
- School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8572 Ibaraki, Japan.
| |
Collapse
|
13
|
Bordukova M, Makarov N, Rodriguez-Esteban R, Schmich F, Menden MP. Generative artificial intelligence empowers digital twins in drug discovery and clinical trials. Expert Opin Drug Discov 2024; 19:33-42. [PMID: 37887266 DOI: 10.1080/17460441.2023.2273839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023]
Abstract
INTRODUCTION The concept of Digital Twins (DTs) translated to drug development and clinical trials describes virtual representations of systems of various complexities, ranging from individual cells to entire humans, and enables in silico simulations and experiments. DTs increase the efficiency of drug discovery and development by digitalizing processes associated with high economic, ethical, or social burden. The impact is multifaceted: DT models sharpen disease understanding, support biomarker discovery and accelerate drug development, thus advancing precision medicine. One way to realize DTs is by generative artificial intelligence (AI), a cutting-edge technology that enables the creation of novel, realistic and complex data with desired properties. AREAS COVERED The authors provide a brief introduction to generative AI and describe how it facilitates the modeling of DTs. In addition, they compare existing implementations of generative AI for DTs in drug discovery and clinical trials. Finally, they discuss technical and regulatory challenges that should be addressed before DTs can transform drug discovery and clinical trials. EXPERT OPINION The current state of DTs in drug discovery and clinical trials does not exploit the entire power of generative AI yet and is limited to simulation of a small number of characteristics. Nonetheless, generative AI has the potential to transform the field by leveraging recent developments in deep learning and customizing models for the needs of scientists, physicians and patients.
Collapse
Affiliation(s)
- Maria Bordukova
- Data & Analytics, Pharmaceutical Research and Early Development, Roche Innovation Center Munich (RICM), Penzberg, Germany
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Munich, Germany
- Department of Biology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Nikita Makarov
- Data & Analytics, Pharmaceutical Research and Early Development, Roche Innovation Center Munich (RICM), Penzberg, Germany
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Munich, Germany
- Department of Biology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Raul Rodriguez-Esteban
- Data & Analytics, Pharmaceutical Research and Early Development, Roche Innovation Center Basel (RICB), Basel, Switzerland
| | - Fabian Schmich
- Data & Analytics, Pharmaceutical Research and Early Development, Roche Innovation Center Munich (RICM), Penzberg, Germany
| | - Michael P Menden
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Munich, Germany
- Department of Biology, Ludwig-Maximilians University Munich, Munich, Germany
- Department of Biochemistry and Pharmacology, University of Melbourne, Melbourne, Australia
- German Center for Diabetes Research (DZD e.V.), Munich, Germany
| |
Collapse
|
14
|
Wistner SC, Rashad L, Slaughter G. Advances in tissue engineering and biofabrication for in vitro skin modeling. BIOPRINTING (AMSTERDAM, NETHERLANDS) 2023; 35:e00306. [PMID: 38645432 PMCID: PMC11031264 DOI: 10.1016/j.bprint.2023.e00306] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The global prevalence of skin disease and injury is continually increasing, yet conventional cell-based models used to study these conditions do not accurately reflect the complexity of human skin. The lack of inadequate in vitro modeling has resulted in reliance on animal-based models to test pharmaceuticals, biomedical devices, and industrial and environmental toxins to address clinical needs. These in vivo models are monetarily and morally expensive and are poor predictors of human tissue responses and clinical trial outcomes. The onset of three-dimensional (3D) culture techniques, such as cell-embedded and decellularized approaches, has offered accessible in vitro alternatives, using innovative scaffolds to improve cell-based models' structural and histological authenticity. However, these models lack adequate organizational control and complexity, resulting in variations between structures and the exclusion of physiologically relevant vascular and immunological features. Recently, biofabrication strategies, which combine biology, engineering, and manufacturing capabilities, have emerged as instrumental tools to recreate the heterogeneity of human skin precisely. Bioprinting uses computer-aided design (CAD) to yield robust and reproducible skin prototypes with unprecedented control over tissue design and assembly. As the interdisciplinary nature of biofabrication grows, we look to the promise of next-generation biofabrication technologies, such as organ-on-a-chip (OOAC) and 4D modeling, to simulate human tissue behaviors more reliably for research, pharmaceutical, and regenerative medicine purposes. This review aims to discuss the barriers to developing clinically relevant skin models, describe the evolution of skin-inspired in vitro structures, analyze the current approaches to biofabricating 3D human skin mimetics, and define the opportunities and challenges in biofabricating skin tissue for preclinical and clinical uses.
Collapse
Affiliation(s)
- Sarah C. Wistner
- Center for Bioelectronics, Old Dominion University, Norfolk, VA, 23508, USA
| | - Layla Rashad
- Center for Bioelectronics, Old Dominion University, Norfolk, VA, 23508, USA
| | - Gymama Slaughter
- Center for Bioelectronics, Old Dominion University, Norfolk, VA, 23508, USA
- Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, VA, 23508, USA
| |
Collapse
|
15
|
Wang N, Zhu S, Lv D, Wang Y, Khawar MB, Sun H. Allosteric modulation of SHP2: Quest from known to unknown. Drug Dev Res 2023; 84:1395-1410. [PMID: 37583266 DOI: 10.1002/ddr.22100] [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: 04/12/2023] [Revised: 07/15/2023] [Accepted: 07/25/2023] [Indexed: 08/17/2023]
Abstract
Src homology-2 domain-containing protein tyrosine phosphatase-2 (SHP2) is a key regulatory factor in the cell cycle and its activating mutations play an important role in the development of various cancers, making it an important target for antitumor drugs. Due to the highly conserved amino acid sequence and positively charged nature of the active site of SHP2, it is difficult to discover inhibitors with high affinity for the catalytic site of SHP2 and sufficient cell permeability, making it considered an "undruggable" target. However, the discovery of allosteric regulation mechanisms provides new opportunities for transforming undruggable targets into druggable ones. Given the limitations of orthosteric inhibitors, SHP2 allosteric inhibitors have become a more selective and safer research direction. In this review, we elucidate the oncogenic mechanism of SHP2 and summarize the discovery methods of SHP2 allosteric inhibitors, providing new strategies for the design and improvement of SHP2 allosteric inhibitors.
Collapse
Affiliation(s)
- Ning Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, China
| | - Shilin Zhu
- Department of Oncology, Haian Hospital of Traditional Chinese Medicine, Haian, China
| | - Dan Lv
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, China
- School of Life Sciences, Anqing Normal University, Anqing, China
| | - Yajun Wang
- Department of Oncology, Haian Hospital of Traditional Chinese Medicine, Haian, China
| | - Muhammad B Khawar
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, China
- Applied Molecular Biology and Biomedicine Lab, Department of Zoology, University of Narowal, Narowal, Pakistan
| | - Haibo Sun
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou, China
| |
Collapse
|
16
|
Abbas ZN, Al-Saffar AZ, Jasim SM, Sulaiman GM. Comparative analysis between 2D and 3D colorectal cancer culture models for insights into cellular morphological and transcriptomic variations. Sci Rep 2023; 13:18380. [PMID: 37884554 PMCID: PMC10603139 DOI: 10.1038/s41598-023-45144-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023] Open
Abstract
Drug development is a time-consuming and expensive process, given the low success rate of clinical trials. Now, anticancer drug developments have shifted to three-dimensional (3D) models which are more likely to mimic tumor behavior compared to traditional two-dimensional (2D) cultures. A comparative study among different aspects was conducted between 2D and 3D cultures using colorectal cancer (CRC) cell lines, in addition, Formalin-Fixed Paraffin-Embedded (FFPE) block samples of patients with CRC were used for evaluation. Compared to the 2D culture, cells grown in 3D displayed significant (p < 0.01) differences in the pattern of cell proliferation over time, cell death phase profile, expression of tumorgenicity-related genes, and responsiveness to 5-fluorouracil, cisplatin, and doxorubicin. Epigenetically, 3D cultures and FFPE shared the same methylation pattern and microRNA expression, while 2D cells showed elevation in methylation rate and altered microRNA expression. Lastly, transcriptomic study depending on RNA sequencing and thorough bioinformatic analyses showed significant (p-adj < 0.05) dissimilarity in gene expression profile between 2D and 3D cultures involving thousands of genes (up/down-regulated) of multiple pathways for each cell line. Taken together, the study provides insights into variations in cellular morphologies between cells cultured in 2D and 3D models.
Collapse
Affiliation(s)
- Zaid Nsaif Abbas
- Department of Molecular and Medical Biotechnology, College of Biotechnology, Al-Nahrain University, Jadriya, Baghdad, Iraq
| | - Ali Z Al-Saffar
- Department of Molecular and Medical Biotechnology, College of Biotechnology, Al-Nahrain University, Jadriya, Baghdad, Iraq.
| | - Saba Mahdi Jasim
- Oncology Teaching Hospital, Medical City, Ministry of Health, Baghdad, Iraq
| | - Ghassan M Sulaiman
- Division of Biotechnology, Department of Applied Sciences, University of Technology, Baghdad, Iraq
| |
Collapse
|
17
|
Domingues C, Jarak I, Veiga F, Dourado M, Figueiras A. Pediatric Drug Development: Reviewing Challenges and Opportunities by Tracking Innovative Therapies. Pharmaceutics 2023; 15:2431. [PMID: 37896191 PMCID: PMC10610377 DOI: 10.3390/pharmaceutics15102431] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/16/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
The paradigm of pediatric drug development has been evolving in a "carrot-and-stick"-based tactic to address population-specific issues. However, the off-label prescription of adult medicines to pediatric patients remains a feature of clinical practice, which may compromise the age-appropriate evaluation of treatments. Therefore, the United States and the European Pediatric Formulation Initiative have recommended applying nanotechnology-based delivery systems to tackle some of these challenges, particularly applying inorganic, polymeric, and lipid-based nanoparticles. Connected with these, advanced therapy medicinal products (ATMPs) have also been highlighted, with optimistic perspectives for the pediatric population. Despite the results achieved using these innovative therapies, a workforce that congregates pediatric patients and/or caregivers, healthcare stakeholders, drug developers, and physicians continues to be of utmost relevance to promote standardized guidelines for pediatric drug development, enabling a fast lab-to-clinical translation. Therefore, taking into consideration the significance of this topic, this work aims to compile the current landscape of pediatric drug development by (1) outlining the historic regulatory panorama, (2) summarizing the challenges in the development of pediatric drug formulation, and (3) delineating the advantages/disadvantages of using innovative approaches, such as nanomedicines and ATMPs in pediatrics. Moreover, some attention will be given to the role of pharmaceutical technologists and developers in conceiving pediatric medicines.
Collapse
Affiliation(s)
- Cátia Domingues
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, 3000-548 Coimbra, Portugal; (C.D.); (I.J.); (F.V.)
- LAQV-REQUIMTE, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Univ Coimbra, Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, 3000-548 Coimbra, Portugal;
| | - Ivana Jarak
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, 3000-548 Coimbra, Portugal; (C.D.); (I.J.); (F.V.)
- Institute for Health Research and Innovation (i3s), University of Porto, 4200-135 Porto, Portugal
| | - Francisco Veiga
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, 3000-548 Coimbra, Portugal; (C.D.); (I.J.); (F.V.)
- LAQV-REQUIMTE, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Marília Dourado
- Univ Coimbra, Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, 3000-548 Coimbra, Portugal;
- Univ Coimbra, Center for Health Studies and Research of the University of Coimbra (CEISUC), Faculty of Medicine, 3000-548 Coimbra, Portugal
- Univ Coimbra, Center for Studies and Development of Continuous and Palliative Care (CEDCCP), Faculty of Medicine, 3000-548 Coimbra, Portugal
| | - Ana Figueiras
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, 3000-548 Coimbra, Portugal; (C.D.); (I.J.); (F.V.)
- LAQV-REQUIMTE, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
18
|
Liu J, Pan Y, Nelson MC, Gooden LK, Metsch LR, Rodriguez AE, Tross S, Del Rio C, Mandler RN, Feaster DJ. Strategies of Managing Repeated Measures: Using Synthetic Random Forest to Predict HIV Viral Suppression Status Among Hospitalized Persons with HIV. AIDS Behav 2023; 27:2915-2931. [PMID: 36739589 PMCID: PMC10403627 DOI: 10.1007/s10461-023-04015-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2023] [Indexed: 02/06/2023]
Abstract
The HIV/AIDS epidemic remains a major public health concern since the 1980s; untreated HIV infection has numerous consequences on quality of life. To optimize patients' health outcomes and to reduce HIV transmission, this study focused on vulnerable populations of people living with HIV (PLWH) and compared different predictive strategies for viral suppression using longitudinal or repeated measures. The four methods of predicting viral suppression are (1) including the repeated measures of each feature as predictors, (2) utilizing only the initial (baseline) value of the feature as predictor, (3) using the last observed value as the predictors and (4) using a growth curve estimated from the features to create individual-specific prediction of growth curves as features. This study suggested the individual-specific prediction of the growth curve performed the best in terms of lowest error rate on an independent set of test data.
Collapse
Affiliation(s)
- Jingxin Liu
- Department of Public Health, Miller School of Medicine, University of Miami, Coral Gables, USA.
- Soffer Clinical Research Ctr, 1120 NW 14th St, Room 1059, Miami, FL, 33136-2107, USA.
| | - Yue Pan
- Department of Public Health, Miller School of Medicine, University of Miami, Coral Gables, USA
| | - Mindy C Nelson
- Department of Public Health, Miller School of Medicine, University of Miami, Coral Gables, USA
| | - Lauren K Gooden
- Department of Sociomedical Sciences, Mailman School of Public Health, Columbia University, New York, USA
| | - Lisa R Metsch
- Department of Sociomedical Sciences, Mailman School of Public Health, Columbia University, New York, USA
| | | | - Susan Tross
- HIV Center for Clinical and Behavioral Studies, New York State Psychiatric Institute and Columbia University Irving Medical Center, New York, NY, USA
| | - Carlos Del Rio
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine and Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Raul N Mandler
- Center for the Clinical Trials Network, National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Daniel J Feaster
- Department of Public Health, Miller School of Medicine, University of Miami, Coral Gables, USA
| |
Collapse
|
19
|
Wang Y, Zhang R, Zhang S, Guo L, Zhou Q, Zhao B, Mo X, Yang Q, Huang Y, Li K, Fan Y, Huang L, Zhou F. OCMR: A comprehensive framework for optical chemical molecular recognition. Comput Biol Med 2023; 163:107187. [PMID: 37393787 DOI: 10.1016/j.compbiomed.2023.107187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/10/2023] [Accepted: 06/19/2023] [Indexed: 07/04/2023]
Abstract
Artificial intelligence (AI) has achieved significant progress in the field of drug discovery. AI-based tools have been used in all aspects of drug discovery, including chemical structure recognition. We propose a chemical structure recognition framework, Optical Chemical Molecular Recognition (OCMR), to improve the data extraction capability in practical scenarios compared with the rule-based and end-to-end deep learning models. The proposed OCMR framework enhances the recognition performances via the integration of local information in the topology of molecular graphs. OCMR handles complex tasks like non-canonical drawing and atomic group abbreviation and substantially improves the current state-of-the-art results on multiple public benchmark datasets and one internally curated dataset.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin, 130012, China; School of Artificial Intelligence, Jilin University, Changchun, 130012, China; College of Computer Science and Technology, Jilin University, Changchun, Jilin, 130012, China
| | - Ruochi Zhang
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin, 130012, China; School of Artificial Intelligence, Jilin University, Changchun, 130012, China
| | - Shengde Zhang
- Machine Learning Department, Silexon AI Technology Co, Ltd, Beijing, 100084, China
| | - Liming Guo
- Machine Learning Department, Silexon AI Technology Co, Ltd, Beijing, 100084, China
| | - Qiong Zhou
- School of Computer Science and Technology, Changchun University of Science and Technology, Changchun, 130012, China
| | - Bowen Zhao
- Machine Learning Department, Silexon AI Technology Co, Ltd, Beijing, 100084, China
| | - Xiaotong Mo
- Machine Learning Department, Silexon AI Technology Co, Ltd, Beijing, 100084, China
| | - Qian Yang
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin, 130012, China; School of Artificial Intelligence, Jilin University, Changchun, 130012, China; College of Computer Science and Technology, Jilin University, Changchun, Jilin, 130012, China
| | - Yajuan Huang
- Machine Learning Department, Silexon AI Technology Co, Ltd, Beijing, 100084, China
| | - Kewei Li
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin, 130012, China; College of Computer Science and Technology, Jilin University, Changchun, Jilin, 130012, China.
| | - Yusi Fan
- College of Software, Jilin University, Changchun, Jilin, 130012, China
| | - Lan Huang
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin, 130012, China; College of Computer Science and Technology, Jilin University, Changchun, Jilin, 130012, China
| | - Fengfeng Zhou
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin, 130012, China; School of Artificial Intelligence, Jilin University, Changchun, 130012, China; College of Computer Science and Technology, Jilin University, Changchun, Jilin, 130012, China.
| |
Collapse
|
20
|
Dalkıran A, Atakan A, Rifaioğlu AS, Martin MJ, Atalay RÇ, Acar AC, Doğan T, Atalay V. Transfer learning for drug-target interaction prediction. Bioinformatics 2023; 39:i103-i110. [PMID: 37387156 DOI: 10.1093/bioinformatics/btad234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2023] [Indexed: 07/01/2023] Open
Abstract
MOTIVATION Utilizing AI-driven approaches for drug-target interaction (DTI) prediction require large volumes of training data which are not available for the majority of target proteins. In this study, we investigate the use of deep transfer learning for the prediction of interactions between drug candidate compounds and understudied target proteins with scarce training data. The idea here is to first train a deep neural network classifier with a generalized source training dataset of large size and then to reuse this pre-trained neural network as an initial configuration for re-training/fine-tuning purposes with a small-sized specialized target training dataset. To explore this idea, we selected six protein families that have critical importance in biomedicine: kinases, G-protein-coupled receptors (GPCRs), ion channels, nuclear receptors, proteases, and transporters. In two independent experiments, the protein families of transporters and nuclear receptors were individually set as the target datasets, while the remaining five families were used as the source datasets. Several size-based target family training datasets were formed in a controlled manner to assess the benefit provided by the transfer learning approach. RESULTS Here, we present a systematic evaluation of our approach by pre-training a feed-forward neural network with source training datasets and applying different modes of transfer learning from the pre-trained source network to a target dataset. The performance of deep transfer learning is evaluated and compared with that of training the same deep neural network from scratch. We found that when the training dataset contains fewer than 100 compounds, transfer learning outperforms the conventional strategy of training the system from scratch, suggesting that transfer learning is advantageous for predicting binders to under-studied targets. AVAILABILITY AND IMPLEMENTATION The source code and datasets are available at https://github.com/cansyl/TransferLearning4DTI. Our web-based service containing the ready-to-use pre-trained models is accessible at https://tl4dti.kansil.org.
Collapse
Affiliation(s)
- Alperen Dalkıran
- Department of Computer Engineering, Middle East Technical University, Ankara 06800, Turkey
- Department of Computer Engineering, Adana Alparslan Türkeş Science and Technology University, Adana 01250, Turkey
| | - Ahmet Atakan
- Department of Computer Engineering, Middle East Technical University, Ankara 06800, Turkey
- Department of Computer Engineering, Erzincan Binali Yıldırım University, Erzincan 24002, Turkey
| | - Ahmet S Rifaioğlu
- Department of Computer Engineering, Iskenderun Technical University, Hatay 31200, Turkey
- Faculty of Medicine, Institute for Computational Biomedicine, Heidelberg University and Heidelberg University Hospital, Heidelberg 69120, Germany
| | - Maria J Martin
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, Hinxton CB10 1SD, United Kingdom
| | - Rengül Çetin Atalay
- Faculty of Pulmonary and Critical Care Medicine, the University of Chicago, Chicago, IL, 60637, United States
| | - Aybar C Acar
- Cancer Systems Biology Laboratory (Kansil), Middle East Technical University, Ankara 06800, Turkey
| | - Tunca Doğan
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, Hinxton CB10 1SD, United Kingdom
- Department of Computer Engineering, Hacettepe University, Ankara 06800, Turkey
| | - Volkan Atalay
- Department of Computer Engineering, Middle East Technical University, Ankara 06800, Turkey
| |
Collapse
|
21
|
Ma S, Alvear A, Schreiner PJ, Seaquist ER, Kirsh T, Chow LS. Development and Validation of an Electronic Health Record-Based Risk Assessment Tool for Hypoglycemia in Patients With Type 2 Diabetes Mellitus. J Diabetes Sci Technol 2023:19322968231184497. [PMID: 37381607 DOI: 10.1177/19322968231184497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
BACKGROUND The recent availability of high-quality data from clinical trials, together with machine learning (ML) techniques, presents exciting opportunities for developing prediction models for clinical outcomes. METHODS As a proof-of-concept, we translated a hypoglycemia risk model derived from the Action to Control Cardiovascular Risk in Diabetes (ACCORD) study into the HypoHazardScore, a risk assessment tool applicable to electronic health record (EHR) data. To assess its performance, we conducted a 16-week clinical study at the University of Minnesota where participants (N = 40) with type 2 diabetes mellitus (T2DM) had hypoglycemia assessed prospectively by continuous glucose monitoring (CGM). RESULTS The HypoHazardScore combines 16 risk factors commonly found within the EHR. The HypoHazardScore successfully predicted (area under the curve [AUC] = 0.723) whether participants experienced least one CGM-assessed hypoglycemic event (glucose <54 mg/dL for ≥15 minutes from two CGMs) while significantly correlating with frequency of CGM-assessed hypoglycemic events (r = 0.38) and percent time experiencing CGM-assessed hypoglycemia (r = 0.39). Compared to participants with a low HypoHazardScore (N = 19, score <4, median score of 4), participants with a high HypoHazardScore (N = 21, score ≥4) had more frequent CGM-assessed hypoglycemic events (high: 1.6 ± 2.2 events/week; low: 0.3 ± 0.5 events/week) and experienced more CGM-assessed hypoglycemia (high: 1.4% ± 2.0%; low: 0.2% ± 0.4% time) during the 16-week follow-up. CONCLUSIONS We demonstrated that a hypoglycemia risk model can be successfully adapted from the ACCORD data to the EHR, with validation by a prospective study using CGM-assessed hypoglycemia. The HypoHazardScore represents a significant advancement toward implementing an EHR-based decision support system that can help reduce hypoglycemia in patients with T2DM.
Collapse
Affiliation(s)
- Sisi Ma
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN, USA
| | - Alison Alvear
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Pamela J Schreiner
- Division of Epidemiology & Community Health, University of Minnesota, Minneapolis, MN, USA
| | | | - Thomas Kirsh
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN, USA
| | - Lisa S Chow
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
22
|
Lin TT, Yang LY, Lin CY, Wang CT, Lai CW, Ko CF, Shih YH, Chen SH. Intelligent De Novo Design of Novel Antimicrobial Peptides against Antibiotic-Resistant Bacteria Strains. Int J Mol Sci 2023; 24:ijms24076788. [PMID: 37047760 PMCID: PMC10095442 DOI: 10.3390/ijms24076788] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/24/2023] [Accepted: 04/02/2023] [Indexed: 04/09/2023] Open
Abstract
Because of the growing number of clinical antibiotic resistance cases in recent years, novel antimicrobial peptides (AMPs) may be ideal for next-generation antibiotics. This study trained a Wasserstein generative adversarial network with gradient penalty (WGAN-GP) based on known AMPs to generate novel AMP candidates. The quality of the GAN-designed peptides was evaluated in silico, and eight of them, named GAN-pep 1–8, were selected by an AMP Artificial Intelligence (AI) classifier and synthesized for further experiments. Disc diffusion testing and minimum inhibitory concentration (MIC) determinations were used to identify the antibacterial effects of the synthesized GAN-designed peptides. Seven of the eight synthesized GAN-designed peptides displayed antibacterial activity. Additionally, GAN-pep 3 and GAN-pep 8 presented a broad spectrum of antibacterial effects and were effective against antibiotic-resistant bacteria strains, such as methicillin-resistant Staphylococcus aureus and carbapenem-resistant Pseudomonas aeruginosa. GAN-pep 3, the most promising GAN-designed peptide candidate, had low MICs against all the tested bacteria. In brief, our approach shows an efficient way to discover AMPs effective against general and antibiotic-resistant bacteria strains. In addition, such a strategy also allows other novel functional peptides to be quickly designed, identified, and synthesized for validation on the wet bench.
Collapse
Affiliation(s)
- Tzu-Tang Lin
- Institute of Information Science, Academia Sinica, Taipei 11529, Taiwan
| | - Li-Yen Yang
- Institute of Information Science, Academia Sinica, Taipei 11529, Taiwan
| | - Chung-Yen Lin
- Institute of Information Science, Academia Sinica, Taipei 11529, Taiwan
| | - Ching-Tien Wang
- Institute of Information Science, Academia Sinica, Taipei 11529, Taiwan
| | - Chia-Wen Lai
- Department of Agricultural Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Chi-Fong Ko
- Department of Agricultural Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Yang-Hsin Shih
- Department of Agricultural Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Shu-Hwa Chen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110301, Taiwan
| |
Collapse
|
23
|
Buckler AJ, Marlevi D, Skenteris NT, Lengquist M, Kronqvist M, Matic L, Hedin U. In silico model of atherosclerosis with individual patient calibration to enable precision medicine for cardiovascular disease. Comput Biol Med 2023; 152:106364. [PMID: 36525832 DOI: 10.1016/j.compbiomed.2022.106364] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/01/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022]
Abstract
OBJECTIVE Guidance for preventing myocardial infarction and ischemic stroke by tailoring treatment for individual patients with atherosclerosis is an unmet need. Such development may be possible with computational modeling. Given the multifactorial biology of atherosclerosis, modeling must be based on complete biological networks that capture protein-protein interactions estimated to drive disease progression. Here, we aimed to develop a clinically relevant scale model of atherosclerosis, calibrate it with individual patient data, and use it to simulate optimized pharmacotherapy for individual patients. APPROACH AND RESULTS The study used a uniquely constituted plaque proteomic dataset to create a comprehensive systems biology disease model for simulating individualized responses to pharmacotherapy. Plaque tissue was collected from 18 patients with 6735 proteins at two locations per patient. 113 pathways were identified and included in the systems biology model of endothelial cells, vascular smooth muscle cells, macrophages, lymphocytes, and the integrated intima, altogether spanning 4411 proteins, demonstrating a range of 39-96% plaque instability. After calibrating the systems biology models for individual patients, we simulated intensive lipid-lowering, anti-inflammatory, and anti-diabetic drugs. We also simulated a combination therapy. Drug response was evaluated as the degree of change in plaque stability, where an improvement was defined as a reduction of plaque instability. In patients with initially unstable lesions, simulated responses varied from high (20%, on combination therapy) to marginal improvement, whereas patients with initially stable plaques showed generally less improvement. CONCLUSION In this pilot study, proteomics-based system biology modeling was shown to simulate drug response based on atherosclerotic plaque instability with a power of 90%, providing a potential strategy for improved personalized management of patients with cardiovascular disease.
Collapse
Affiliation(s)
- Andrew J Buckler
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Elucid Bioimaging Inc., Boston, MA, USA
| | - David Marlevi
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Nikolaos T Skenteris
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Mariette Lengquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Malin Kronqvist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
24
|
Sousa AS, Serra J, Estevens C, Costa R, Ribeiro AJ. A quality by design approach in oral extended release drug delivery systems: where we are and where we are going? JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022. [DOI: 10.1007/s40005-022-00603-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
25
|
Pandiyan S, Wang L. A comprehensive review on recent approaches for cancer drug discovery associated with artificial intelligence. Comput Biol Med 2022; 150:106140. [PMID: 36179510 DOI: 10.1016/j.compbiomed.2022.106140] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 07/20/2022] [Accepted: 09/18/2022] [Indexed: 11/03/2022]
Abstract
Through the revolutionization of artificial intelligence (AI) technologies in clinical research, significant improvement is observed in diagnosis of cancer. Utilization of these AI technologies, such as machine and deep learning, is imperative for the discovery of novel anticancer drugs and improves existing/ongoing cancer therapeutics. However, building a model for complicated cancers and their types remains a challenge due to lack of effective therapeutics that hinder the establishment of effective computational tools. In this review, we exploit recent approaches and state-of-the-art in implementing AI methods for anticancer drug discovery, and discussed how advances in these applications need to be considered in the current cancer therapeutics. Considering the immense potential of AI, we explore molecular docking and their interactions to recognize metabolic activities that support drug design. Finally, we highlight corresponding strategies in applying machine and deep learning methods to various types of cancer with their pros and cons.
Collapse
Affiliation(s)
- Sanjeevi Pandiyan
- Research Center for Intelligent Information Technology, Nantong University, Nantong, China; School of Information Science and Technology, Nantong University, Nantong, China; Nantong Research Institute for Advanced Communication Technologies, Nantong, China
| | - Li Wang
- Research Center for Intelligent Information Technology, Nantong University, Nantong, China; School of Information Science and Technology, Nantong University, Nantong, China; Nantong Research Institute for Advanced Communication Technologies, Nantong, China.
| |
Collapse
|
26
|
Stepanov D, Buchmann D, Schultze N, Wolber G, Schaufler K, Guenther S, Belik V. A Combined Bayesian and Similarity-Based Approach for Predicting E. coli Biofilm Inhibition by Phenolic Natural Compounds. JOURNAL OF NATURAL PRODUCTS 2022; 85:2255-2265. [PMID: 36107719 DOI: 10.1021/acs.jnatprod.2c00005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Screening for biofilm inhibition by purified natural compounds is difficult due to compounds' chemical diversity and limited commercial availability, combined with time- and cost-intensiveness of the laboratory process. In silico prediction of chemical and biological properties of molecules is a widely used technique when experimental data availability is of concern. At the same time, the performance of predictive models directly depends on the amount and quality of experimental data. Driven by the interest in developing a model for prediction of the antibiofilm effect of phenolic natural compounds such as flavonoids, we performed experimental assessment of antibiofilm activity of 320 compounds from this subset of chemicals. The assay was performed once on two Escherichia coli strains on agar in 24-well microtiter plates. The inhibition was assessed visually by detecting morphological changes in macrocolonies. Using the data obtained, we subsequently trained a Bayesian logistic regression model for prediction of biofilm inhibition, which was combined with a similarity-based method in order to increase the overall sensitivity (at the cost of accuracy). The quality of the predictions was subsequently validated by experimental assessment in three independent experiments with two resistant E. coli strains of 23 compounds absent in the initial data set. The validation demonstrated that the model may successfully predict the targeted effect as compared to the baseline accuracy. Using a randomly selected database of commercially available natural phenolics, we obtained approximately 6.0% of active compounds, whereas using our prediction-based substance selection, the percentage of phenolics found to be active increased to 34.8%.
Collapse
Affiliation(s)
- Dmitri Stepanov
- Department of Biology, Chemistry & Pharmacy, Institute of Pharmacy, Pharmaceutical and Medicinal Chemistry, Freie Universität Berlin, Königin-Luise-Straße 2+4, 14195 Berlin, Germany
| | - David Buchmann
- Pharmaceutical Biology, Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489 Greifswald, Germany
| | - Nadin Schultze
- Pharmaceutical Biology, Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489 Greifswald, Germany
| | - Gerhard Wolber
- Department of Biology, Chemistry & Pharmacy, Institute of Pharmacy, Pharmaceutical and Medicinal Chemistry, Freie Universität Berlin, Königin-Luise-Straße 2+4, 14195 Berlin, Germany
| | - Katharina Schaufler
- Pharmaceutical Microbiology, Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489 Greifswald, Germany
- Institute of Infection Medicine, Christian-Albrecht University Kiel and University Medical Center Schleswig-Holstein, 24105 Kiel, Germany
| | - Sebastian Guenther
- Pharmaceutical Biology, Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489 Greifswald, Germany
| | - Vitaly Belik
- Department of Veterinary Medicine, Institute for Veterinary Epidemiology and Biostatistics, System Modeling Group, Freie Universität Berlin, Königsweg 67, 14163 Berlin, Germany
| |
Collapse
|
27
|
Wang FS, Chen PR, Chen TY, Zhang HX. Fuzzy optimization for identifying anti-cancer targets with few side effects in constraint-based models of head and neck cancer. ROYAL SOCIETY OPEN SCIENCE 2022; 9:220633. [PMID: 36303939 PMCID: PMC9597175 DOI: 10.1098/rsos.220633] [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: 05/26/2022] [Accepted: 09/27/2022] [Indexed: 06/16/2023]
Abstract
Computer-aided methods can be used to screen potential candidate targets and to reduce the time and cost of drug development. In most of these methods, synthetic lethality is used as a therapeutic criterion to identify drug targets. However, these methods do not consider the side effects during the identification stage. This study developed a fuzzy multi-objective optimization for identifying anti-cancer targets that not only evaluated cancer cell mortality, but also minimized side effects due to treatment. We identified potential anti-cancer enzymes and antimetabolites for the treatment of head and neck cancer (HNC). The identified one- and two-target enzymes were primarily involved in six major pathways, namely, purine and pyrimidine metabolism and the pentose phosphate pathway. Most of the identified targets can be regulated by approved drugs; thus, these drugs are potential candidates for drug repurposing as a treatment for HNC. Furthermore, we identified antimetabolites involved in pathways similar to those identified using a gene-centric approach. Moreover, HMGCR knockdown could not block the growth of HNC cells. However, the two-target combinations of (UMPS, HMGCR) and (CAD, HMGCR) could achieve cell mortality and improve metabolic deviation grades over 22% without reducing the cell viability grade.
Collapse
Affiliation(s)
- Feng-Sheng Wang
- Department of Chemical Engineering, National Chung Cheng University, Chiayi, Taiwan
| | - Pei-Rong Chen
- Department of Chemical Engineering, National Chung Cheng University, Chiayi, Taiwan
| | - Ting-Yu Chen
- Department of Chemical Engineering, National Chung Cheng University, Chiayi, Taiwan
| | - Hao-Xiang Zhang
- Department of Chemical Engineering, National Chung Cheng University, Chiayi, Taiwan
| |
Collapse
|
28
|
Hadiby S, Ali YMB. FNN Based-Virtual Screening Using 2D Pharmacophore Fingerprint for Activity Prediction in Drug Discovery. INTERNATIONAL JOURNAL OF COMPUTATIONAL INTELLIGENCE AND APPLICATIONS 2022. [DOI: 10.1142/s1469026822500195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Drug discovery remains a hard field that faces from the beginning of its process to the end many difficulties and challenges in order to discover a new potential drug. The use of technology has helped a lot in achieving many goals at the lowest cost and in the shortest possible time. Machine learning methods have proven for many years their performance although their limitations in some cases. The use of deep learning for virtual screening in drug discovery allows to process efficiently the huge amount of data and gives more precise results. In this paper, we propose a procedure for virtual screening (VS) based on Feedforward Neural Network in order to predict the biological activity of a set of chemical compounds on a given receptor. we have proposed a distance interval and it divisions to describe the chemical compound by the 2D pharmacophore fingerprint. Our model was trained on a dataset of active and inactive chemical compounds on cyclin A kinase1 receptor (CDK1), a very important protein family which has a role in the regulation of the cell cycle and cancer development. The results have proven that the proposed model is efficient and comparable with some widely used machine learning methods in drug discovery.
Collapse
Affiliation(s)
- Seloua Hadiby
- Department of Computer Science, Computer Research Laboratory, Badji Mokhtar University, Annaba, Algeria
| | - Yamina Mohamed Ben Ali
- Department of Computer Science, Computer Research Laboratory, Badji Mokhtar University, Annaba, Algeria
| |
Collapse
|
29
|
Herrera-Bravo J, Farías JG, Sandoval C, Herrera-Belén L, Quiñones J, Díaz R, Beltrán JF. nAChR-PEP-PRED: A Robust Tool for Predicting Peptide Inhibitors of Acetylcholine Receptors Using the Random Forest Classifier. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10460-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
30
|
Feasibility and application of machine learning enabled fast screening of poly-beta-amino-esters for cartilage therapies. Sci Rep 2022; 12:14215. [PMID: 35987777 PMCID: PMC9392801 DOI: 10.1038/s41598-022-18332-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Despite the large prevalence of diseases affecting cartilage (e.g. knee osteoarthritis affecting 16% of population globally), no curative treatments are available because of the limited capacity of drugs to localise in such tissue caused by low vascularisation and electrostatic repulsion. While an effective delivery system is sought, the only option is using high drug doses that can lead to systemic side effects. We introduced poly-beta-amino-esters (PBAEs) to effectively deliver drugs into cartilage tissues. PBAEs are copolymer of amines and di-acrylates further end-capped with other amine; therefore encompassing a very large research space for the identification of optimal candidates. In order to accelerate the screening of all possible PBAEs, the results of a small pool of polymers (n = 90) were used to train a variety of machine learning (ML) methods using only polymers properties available in public libraries or estimated from the chemical structure. Bagged multivariate adaptive regression splines (MARS) returned the best predictive performance and was used on the remaining (n = 3915) possible PBAEs resulting in the recognition of pivotal features; a further round of screening was carried out on PBAEs (n = 150) with small variations of structure of the main candidates from the first round. The refinements of such characteristics enabled the identification of a leading candidate predicted to improve drug uptake > 20 folds over conventional clinical treatment; this uptake improvement was also experimentally confirmed. This work highlights the potential of ML to accelerate biomaterials development by efficiently extracting information from a limited experimental dataset thus allowing patients to benefit earlier from a new technology and at a lower price. Such roadmap could also be applied for other drug/materials development where optimisation would normally be approached through combinatorial chemistry.
Collapse
|
31
|
Ono T, Denda R, Tsukahara Y, Nakamura T, Okamoto K, Takayanagi H, Nakashima T. Simultaneous augmentation of muscle and bone by locomomimetism through calcium-PGC-1α signaling. Bone Res 2022; 10:52. [PMID: 35918335 PMCID: PMC9345981 DOI: 10.1038/s41413-022-00225-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 06/01/2022] [Accepted: 06/19/2022] [Indexed: 11/09/2022] Open
Abstract
Impaired locomotion has been extensively studied worldwide because those afflicted with it have a potential risk of becoming bedridden. Physical exercise at times can be an effective remedy for frailty, but exercise therapy cannot be applied in all clinical cases. Medication is safer than exercise, but there are no drugs that reinforce both muscle and bone when administered alone. Multiple medications increase the risk of adverse events; thus, there is a need for individual drugs targeting both tissues. To this end, we established a novel sequential drug screening system and identified an aminoindazole derivative, locamidazole (LAMZ), which promotes both myogenesis and osteoblastogenesis while suppressing osteoclastogenesis. Administration of this drug enhanced locomotor function, with muscle and bone significantly strengthened. Mechanistically, LAMZ induced Mef2c and PGC-1α in a calcium signaling-dependent manner. As this signaling is activated upon physical exercise, LAMZ mimics physical exercise. Thus, LAMZ is a promising therapeutic drug for locomotor diseases, including sarcopenia and osteoporosis.
Collapse
Affiliation(s)
- Takehito Ono
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Ryosuke Denda
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Yuta Tsukahara
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan
- School of Dentistry, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Takashi Nakamura
- Department of Biochemistry, Tokyo Dental College, Kandamisakicho 2-9-18, Chiyoda-ku, Tokyo, 101-0061, Japan
| | - Kazuo Okamoto
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Tomoki Nakashima
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan.
| |
Collapse
|
32
|
The future of Artificial Intelligence for the BioTech Big Data landscape. Curr Opin Biotechnol 2022; 76:102714. [DOI: 10.1016/j.copbio.2022.102714] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/17/2022] [Accepted: 03/02/2022] [Indexed: 01/17/2023]
|
33
|
Keutzer L, You H, Farnoud A, Nyberg J, Wicha SG, Maher-Edwards G, Vlasakakis G, Moghaddam GK, Svensson EM, Menden MP, Simonsson USH. Machine Learning and Pharmacometrics for Prediction of Pharmacokinetic Data: Differences, Similarities and Challenges Illustrated with Rifampicin. Pharmaceutics 2022; 14:pharmaceutics14081530. [PMID: 35893785 PMCID: PMC9330804 DOI: 10.3390/pharmaceutics14081530] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 01/27/2023] Open
Abstract
Pharmacometrics (PM) and machine learning (ML) are both valuable for drug development to characterize pharmacokinetics (PK) and pharmacodynamics (PD). Pharmacokinetic/pharmacodynamic (PKPD) analysis using PM provides mechanistic insight into biological processes but is time- and labor-intensive. In contrast, ML models are much quicker trained, but offer less mechanistic insights. The opportunity of using ML predictions of drug PK as input for a PKPD model could strongly accelerate analysis efforts. Here exemplified by rifampicin, a widely used antibiotic, we explore the ability of different ML algorithms to predict drug PK. Based on simulated data, we trained linear regressions (LASSO), Gradient Boosting Machines, XGBoost and Random Forest to predict the plasma concentration-time series and rifampicin area under the concentration-versus-time curve from 0–24 h (AUC0–24h) after repeated dosing. XGBoost performed best for prediction of the entire PK series (R2: 0.84, root mean square error (RMSE): 6.9 mg/L, mean absolute error (MAE): 4.0 mg/L) for the scenario with the largest data size. For AUC0–24h prediction, LASSO showed the highest performance (R2: 0.97, RMSE: 29.1 h·mg/L, MAE: 18.8 h·mg/L). Increasing the number of plasma concentrations per patient (0, 2 or 6 concentrations per occasion) improved model performance. For example, for AUC0–24h prediction using LASSO, the R2 was 0.41, 0.69 and 0.97 when using predictors only (no plasma concentrations), 2 or 6 plasma concentrations per occasion as input, respectively. Run times for the ML models ranged from 1.0 s to 8 min, while the run time for the PM model was more than 3 h. Furthermore, building a PM model is more time- and labor-intensive compared with ML. ML predictions of drug PK could thus be used as input into a PKPD model, enabling time-efficient analysis.
Collapse
Affiliation(s)
- Lina Keutzer
- Department of Pharmaceutical Biosciences, Uppsala University, 75124 Uppsala, Sweden; (L.K.); (H.Y.)
| | - Huifang You
- Department of Pharmaceutical Biosciences, Uppsala University, 75124 Uppsala, Sweden; (L.K.); (H.Y.)
| | - Ali Farnoud
- Computational Health Center, Helmholtz Munich, 85764 Neuherberg, Germany; (A.F.); (M.P.M.)
| | - Joakim Nyberg
- Department of Pharmacy, Uppsala University, 75123 Uppsala, Sweden; (J.N.); (E.M.S.)
| | - Sebastian G. Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, 20146 Hamburg, Germany;
| | - Gareth Maher-Edwards
- Research, Clinical Pharmacology Modelling & Simulation, GlaxoSmithKline, London TW8 9GS, UK; (G.M.-E.); (G.V.); (G.K.M.)
| | - Georgios Vlasakakis
- Research, Clinical Pharmacology Modelling & Simulation, GlaxoSmithKline, London TW8 9GS, UK; (G.M.-E.); (G.V.); (G.K.M.)
| | - Gita Khalili Moghaddam
- Research, Clinical Pharmacology Modelling & Simulation, GlaxoSmithKline, London TW8 9GS, UK; (G.M.-E.); (G.V.); (G.K.M.)
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Elin M. Svensson
- Department of Pharmacy, Uppsala University, 75123 Uppsala, Sweden; (J.N.); (E.M.S.)
- Department of Pharmacy, Radboud Institute of Health Sciences, Radboud University Medical Center, 6525 EZ Nijmegen, The Netherlands
| | - Michael P. Menden
- Computational Health Center, Helmholtz Munich, 85764 Neuherberg, Germany; (A.F.); (M.P.M.)
- Department of Biology, Ludwig-Maximilian University Munich, 82152 Planegg-Martinsried, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
| | - Ulrika S. H. Simonsson
- Department of Pharmaceutical Biosciences, Uppsala University, 75124 Uppsala, Sweden; (L.K.); (H.Y.)
- Correspondence:
| | | |
Collapse
|
34
|
Jung YL, Yoo HS, Hwang J. Artificial intelligence-based decision support model for new drug development planning. EXPERT SYSTEMS WITH APPLICATIONS 2022; 198:116825. [PMID: 35283560 PMCID: PMC8902892 DOI: 10.1016/j.eswa.2022.116825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 01/18/2022] [Accepted: 03/02/2022] [Indexed: 06/14/2023]
Abstract
New drug development guarantees a very high return on success, but the success rate is extremely low. Pharmaceutical companies have attempted to use various strategies to increase the success rate of drug development, but this goal has been difficult to achieve. In this study, we developed a model that can guide effective decision-making at the planning stage of new drug development by leveraging machine learning. The Drug Development Recommendation (DDR) model, we present here, is a hybrid model for recommending and/or predicting drug groups suitable for development by individual pharmaceutical companies. It combines association rule learning, collaborative filtering, and content-based filtering approaches for enterprise-customized recommendations. In the case of content-based filtering applying a random forest classification algorithm, the accuracy and area under curve were 78% and 0.74, respectively. In particular, the DDR model was applied to predict the success probability of companies developing Coronavirus disease 2019 (COVID-19) vaccines. It was demonstrated that the higher the predicted score from the DDR model, the more progress in the clinical phase of the COVID-19 vaccine development. Although our approach has limitations that should be improved, it makes scientific as well as industrial contributions in that the DDR model can support rational decision-making prior to initiating drug development by considering not only technical aspects but also company-related variables.
Collapse
Affiliation(s)
- Ye Lim Jung
- Division of Data Analysis, Korea Institute of Science and Technology Information (KISTI), Seoul 02456, Republic of Korea
| | - Hyoung Sun Yoo
- Division of Data Analysis, Korea Institute of Science and Technology Information (KISTI), Seoul 02456, Republic of Korea
- Science and Technology Management Policy, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - JeeNa Hwang
- Division of Data Analysis, Korea Institute of Science and Technology Information (KISTI), Seoul 02456, Republic of Korea
| |
Collapse
|
35
|
Diagnosis and prognosis of COVID-19 employing analysis of patients' plasma and serum via LC-MS and machine learning. Comput Biol Med 2022; 146:105659. [PMID: 35751188 PMCID: PMC9123826 DOI: 10.1016/j.compbiomed.2022.105659] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 05/11/2022] [Accepted: 05/18/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE To implement and evaluate machine learning (ML) algorithms for the prediction of COVID-19 diagnosis, severity, and fatality and to assess biomarkers potentially associated with these outcomes. MATERIAL AND METHODS Serum (n = 96) and plasma (n = 96) samples from patients with COVID-19 (acute, severe and fatal illness) from two independent hospitals in China were analyzed by LC-MS. Samples from healthy volunteers and from patients with pneumonia caused by other viruses (i.e. negative RT-PCR for COVID-19) were used as controls. Seven different ML-based models were built: PLS-DA, ANNDA, XGBoostDA, SIMCA, SVM, LREG and KNN. RESULTS The PLS-DA model presented the best performance for both datasets, with accuracy rates to predict the diagnosis, severity and fatality of COVID-19 of 93%, 94% and 97%, respectively. Low levels of the metabolites ribothymidine, 4-hydroxyphenylacetoylcarnitine and uridine were associated with COVID-19 positivity, whereas high levels of N-acetyl-glucosamine-1-phosphate, cysteinylglycine, methyl isobutyrate, l-ornithine and 5,6-dihydro-5-methyluracil were significantly related to greater severity and fatality from COVID-19. CONCLUSION The PLS-DA model can help to predict SARS-CoV-2 diagnosis, severity and fatality in daily practice. Some biomarkers typically increased in COVID-19 patients' serum or plasma (i.e. ribothymidine, N-acetyl-glucosamine-1-phosphate, l-ornithine, 5,6-dihydro-5-methyluracil) should be further evaluated as prognostic indicators of the disease.
Collapse
|
36
|
Dutta A. Predicting Drug Mechanics by Deep Learning on Gene and Cell Activities. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2022; 2022:2916-2919. [PMID: 36086025 DOI: 10.1109/embc48229.2022.9871391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The identification of protein target and mechanism of disease are fundamentally important in drug discovery. A pipeline for predicting mechanism of action (MoA) for drug molecules based on gene expression and cell viability data is developed and demonstrated on experimental data. A deep learning network learns the known MoAs over thousands of gene expression and cell viability training data and is shown to predict the unknown MoAs over test data with high efficacy.
Collapse
|
37
|
Kaushal K, Sarma P, Rana SV, Medhi B, Naithani M. Emerging role of artificial intelligence in therapeutics for COVID-19: a systematic review. J Biomol Struct Dyn 2022; 40:4750-4765. [PMID: 33300456 PMCID: PMC7738208 DOI: 10.1080/07391102.2020.1855250] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/20/2020] [Indexed: 12/21/2022]
Abstract
To elucidate the role of artificial intelligence (AI) in therapeutics for coronavirus disease 2019 (COVID-19). Five databases were searched (December 2019-May 2020). We included both published and pre-print original articles in English that applied AI, machine learning or deep learning in drug repurposing, novel drug discovery, vaccine and antibody development for COVID-19. Out of 31 studies included, 16 studies applied AI for drug repurposing, whereas 10 studies utilized AI for novel drug discovery. Only four studies used AI technology for vaccine development, whereas one study generated stable antibodies against SARS-CoV-2. Approx. 50% of studies exclusively targeted 3CLpro of SARS-CoV-2, and only two studies targeted ACE/TMPSS2 for inhibiting host viral interactions. Around 16% of the identified drugs are in different phases of clinical evaluation against COVID-19. AI has emerged as a promising solution of COVID-19 therapeutics. During this current pandemic, many of the researchers have used AI-based strategies to process large databases in a more customized manner leading to the faster identification of several potential targets, novel/repurposing of drugs and vaccine candidates. A number of these drugs are either approved or are in a late-stage clinical trial and are potentially effective against SARS-CoV2 indicating validity of the methodology. However, as the use of AI-based screening program is currently in budding stage, sole reliance on such algorithms is not advisable at this current point of time and an evidence based approach is warranted to confirm their usefulness against this life-threatening disease. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Karanvir Kaushal
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh, India
| | - Phulan Sarma
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - S. V. Rana
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Manisha Naithani
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh, India
| |
Collapse
|
38
|
Sampaio PNS, Calado CRC. Antimicrobial evaluation of the Cynara cardunculus extract in Helicobacter pylori cells using mid-infrared spectroscopy and chemometric methods. J Appl Microbiol 2022; 133:1743-1756. [PMID: 35729780 DOI: 10.1111/jam.15679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 05/17/2022] [Accepted: 06/19/2022] [Indexed: 11/29/2022]
Abstract
AIMS The treatment effectiveness of gastric diseases caused by the bacteria Helicobacter pylori is failing due to high resistance to some antibiotics. Consequently, it is urgent to develop an accurate methodology to screen new antimicrobial agents. METHODS AND RESULTS A preliminary assay, using both therapeutic-based antibiotics (clarithromycin and metronidazole), was conducted to optimize experimental conditions in terms of the sensibility of the Fourier Transform Mid-Infrared spectroscopy (MIR-FTIR) associated with chemometric methods. Principal component analysis was applied to understand how the Cynara extract concentration acts differentially against H. pylori bacteria. The partial least squares model, characterized by R2 = 0.98, and root mean square error cross-validation, 0.011, was developed for the spectral regions (3600 - 2500 cm-1 , and 2000 - 698 cm-1 ). CONCLUSIONS MIR-FTIR spectroscopy associated with chemometric methods can be considered a suitable approach to discover and analyze the promissory antimicrobial agents based on the biomolecular changes observed according to the Cynara extract. SIGNIFICANCE AND IMPACT OF THE STUDY MIR-FTIR spectroscopy and chemometric methods allowed to register the biomolecular changes due to the potential antimicrobial drugs at reduced concentrations comparatively to the conventional assay based on an agar-dilution method, being considered a useful approach to develop a platform to discover new bioactive molecules, allowing to reduce time and costs related to the exploratory step.
Collapse
Affiliation(s)
- Pedro N Sousa Sampaio
- DREAMS-Centre for Interdisciplinary Development and Research on Environment, Applied Management and Space, Faculty of Engineering, Lusófona University (ULHT), Lisbon, Portugal
| | - Cecília R C Calado
- CIMOSM - Centro de Investigação em Modelação e Optimização de Sistemas Multifuncionais, Instituto Superior de Engenharia de Lisboa (ISEL), Instituto Politécnico de Lisboa Rua Conselheiro Emídio Navarro, 1, 1959-007, Lisbon, Portugal
| |
Collapse
|
39
|
Nagy B, Galata DL, Farkas A, Nagy ZK. Application of Artificial Neural Networks in the Process Analytical Technology of Pharmaceutical Manufacturing-a Review. AAPS J 2022; 24:74. [PMID: 35697951 DOI: 10.1208/s12248-022-00706-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/06/2022] [Indexed: 01/22/2023] Open
Abstract
Industry 4.0 has started to transform the manufacturing industries by embracing digitalization, automation, and big data, aiming for interconnected systems, autonomous decisions, and smart factories. Machine learning techniques, such as artificial neural networks (ANN), have emerged as potent tools to address the related computational tasks. These advancements have also reached the pharmaceutical industry, where the Process Analytical Technology (PAT) initiative has already paved the way for the real-time analysis of the processes and the science- and risk-based flexible production. This paper aims to assess the potential of ANNs within the PAT concept to aid the modernization of pharmaceutical manufacturing. The current state of ANNs is systematically reviewed for the most common manufacturing steps of solid pharmaceutical products, and possible research gaps and future directions are identified. In this way, this review could aid the further development of machine learning techniques for pharmaceutical production and eventually contribute to the implementation of intelligent manufacturing lines with automated quality assurance.
Collapse
Affiliation(s)
- Brigitta Nagy
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., Budapest, H-1111, Hungary
| | - Dorián László Galata
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., Budapest, H-1111, Hungary
| | - Attila Farkas
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., Budapest, H-1111, Hungary
| | - Zsombor Kristóf Nagy
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3., Budapest, H-1111, Hungary.
| |
Collapse
|
40
|
Lo-Thong-Viramoutou O, Charton P, Cadet XF, Grondin-Perez B, Saavedra E, Damour C, Cadet F. Non-linearity of Metabolic Pathways Critically Influences the Choice of Machine Learning Model. Front Artif Intell 2022; 5:744755. [PMID: 35757298 PMCID: PMC9226554 DOI: 10.3389/frai.2022.744755] [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] [Received: 07/20/2021] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
The use of machine learning (ML) in life sciences has gained wide interest over the past years, as it speeds up the development of high performing models. Important modeling tools in biology have proven their worth for pathway design, such as mechanistic models and metabolic networks, as they allow better understanding of mechanisms involved in the functioning of organisms. However, little has been done on the use of ML to model metabolic pathways, and the degree of non-linearity associated with them is not clear. Here, we report the construction of different metabolic pathways with several linear and non-linear ML models. Different types of data are used; they lead to the prediction of important biological data, such as pathway flux and final product concentration. A comparison reveals that the data features impact model performance and highlight the effectiveness of non-linear models (e.g., QRF: RMSE = 0.021 nmol·min-1 and R2 = 1 vs. Bayesian GLM: RMSE = 1.379 nmol·min-1 R2 = 0.823). It turns out that the greater the degree of non-linearity of the pathway, the better suited a non-linear model will be. Therefore, a decision-making support for pathway modeling is established. These findings generally support the hypothesis that non-linear aspects predominate within the metabolic pathways. This must be taken into account when devising possible applications of these pathways for the identification of biomarkers of diseases (e.g., infections, cancer, neurodegenerative diseases) or the optimization of industrial production processes.
Collapse
Affiliation(s)
- Ophélie Lo-Thong-Viramoutou
- University of Paris, BIGR—Biologie Intégrée du Globule Rouge, Inserm, UMR_S1134, Paris, France
- Laboratory of Excellence GR-Ex, Paris, France
- Laboratory DSIMB, UMR_S1134, BIGR, Inserm, Faculty of Sciences and Technology, University of La Reunion, Saint-Denis, France
| | - Philippe Charton
- University of Paris, BIGR—Biologie Intégrée du Globule Rouge, Inserm, UMR_S1134, Paris, France
- Laboratory of Excellence GR-Ex, Paris, France
- Laboratory DSIMB, UMR_S1134, BIGR, Inserm, Faculty of Sciences and Technology, University of La Reunion, Saint-Denis, France
| | | | - Brigitte Grondin-Perez
- EnergyLab, EA 4079, Faculty of Sciences and Technology, University of La Reunion, Saint-Denis, France
| | - Emma Saavedra
- Departamento de Bioquímica, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Cédric Damour
- EnergyLab, EA 4079, Faculty of Sciences and Technology, University of La Reunion, Saint-Denis, France
| | - Frédéric Cadet
- University of Paris, BIGR—Biologie Intégrée du Globule Rouge, Inserm, UMR_S1134, Paris, France
- Laboratory of Excellence GR-Ex, Paris, France
- Laboratory DSIMB, UMR_S1134, BIGR, Inserm, Faculty of Sciences and Technology, University of La Reunion, Saint-Denis, France
| |
Collapse
|
41
|
Machine learning modeling for the prediction of materials energy. Neural Comput Appl 2022. [DOI: 10.1007/s00521-022-07416-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
42
|
Deep Learning Based-Virtual Screening Using 2D Pharmacophore Fingerprint in Drug Discovery. Neural Process Lett 2022. [DOI: 10.1007/s11063-022-10879-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
43
|
Puzzle Out Machine Learning Model-Explaining Disintegration Process in ODTs. Pharmaceutics 2022; 14:pharmaceutics14040859. [PMID: 35456693 PMCID: PMC9044744 DOI: 10.3390/pharmaceutics14040859] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/10/2022] [Accepted: 04/11/2022] [Indexed: 02/05/2023] Open
Abstract
Tablets are the most common dosage form of pharmaceutical products. While tablets represent the majority of marketed pharmaceutical products, there remain a significant number of patients who find it difficult to swallow conventional tablets. Such difficulties lead to reduced patient compliance. Orally disintegrating tablets (ODT), sometimes called oral dispersible tablets, are the dosage form of choice for patients with swallowing difficulties. ODTs are defined as a solid dosage form for rapid disintegration prior to swallowing. The disintegration time, therefore, is one of the most important and optimizable critical quality attributes (CQAs) for ODTs. Current strategies to optimize ODT disintegration times are based on a conventional trial-and-error method whereby a small number of samples are used as proxies for the compliance of whole batches. We present an alternative machine learning approach to optimize the disintegration time based on a wide variety of machine learning (ML) models through the H2O AutoML platform. ML models are presented with inputs from a database originally presented by Han et al., which was enhanced and curated to include chemical descriptors representing active pharmaceutical ingredient (API) characteristics. A deep learning model with a 10-fold cross-validation NRMSE of 8.1% and an R2 of 0.84 was obtained. The critical parameters influencing the disintegration of the directly compressed ODTs were ascertained using the SHAP method to explain ML model predictions. A reusable, open-source tool, the ODT calculator, is now available at Heroku platform.
Collapse
|
44
|
Pillai N, Dasgupta A, Sudsakorn S, Fretland J, Mavroudis PD. Machine-learning-guided early drug discovery of small molecules. Drug Discov Today 2022; 27:2209-2215. [PMID: 35364270 DOI: 10.1016/j.drudis.2022.03.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 12/19/2021] [Accepted: 03/25/2022] [Indexed: 11/17/2022]
Abstract
Machine learning (ML) approaches have been widely adopted within the early stages of the drug discovery process, particularly within the context of small-molecule drug candidates. Despite this, the use of ML is still limited in the pharmacokinetic/pharmacodynamic (PK/PD) application space. Here, we describe recent progress and the role of ML used in preclinical drug discovery. We summarize the advances and current strategies used to predict ADME properties of small molecules based on their structures, and predict structures based on the desired properties for molecular screening and optimization. Finally, we discuss the use of ML to predict PK to rank the ability of drug candidates to achieve appropriate exposures and hence provide important insights into safety and efficacy. Teaser: In this short review we highlight key advances in the application of machine learning to early drug discovery for small molecules in the preclinical setting.
Collapse
Affiliation(s)
- Nikhil Pillai
- Quantitative Pharmacology, DMPK, Sanofi US, Waltham, MA, USA.
| | - Aparajita Dasgupta
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | | | |
Collapse
|
45
|
Staszak M, Staszak K, Wieszczycka K, Bajek A, Roszkowski K, Tylkowski B. Machine learning in drug design: Use of artificial intelligence to explore the chemical structure–biological activity relationship. WIRES COMPUTATIONAL MOLECULAR SCIENCE 2022. [DOI: 10.1002/wcms.1568] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Maciej Staszak
- Institute of Technology and Chemical Engineering Poznan University of Technology Poznan Poland
| | - Katarzyna Staszak
- Institute of Technology and Chemical Engineering Poznan University of Technology Poznan Poland
| | - Karolina Wieszczycka
- Institute of Technology and Chemical Engineering Poznan University of Technology Poznan Poland
| | - Anna Bajek
- Department of Tissue Engineering Collegium Medicum, Nicolaus Copernicus University Bydgoszcz Poland
| | - Krzysztof Roszkowski
- Department of Oncology Collegium Medicum Nicolaus Copernicus University Bydgoszcz Poland
| | - Bartosz Tylkowski
- Department of Chemical Engineering University Rovira i Virgili Tarragona Spain
- Eurecat, Centre Tecnològic de Catalunya Chemical Technologies Unit Tarragona Spain
| |
Collapse
|
46
|
Yoo S, Kim J, Choi GJ. Drug Properties Prediction Based on Deep Learning. Pharmaceutics 2022; 14:467. [PMID: 35214201 PMCID: PMC8880629 DOI: 10.3390/pharmaceutics14020467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 01/31/2023] Open
Abstract
In recent research on the formulation prediction of oral dissolving drugs, deep learning models with significantly improved performance compared to machine learning models were proposed. However, the performance degradation due to limitations of an imbalanced dataset with a small size and inefficient neural network structure has still not been resolved. Therefore, we propose new deep learning-based prediction models that maximize the prediction performance for disintegration time of oral fast disintegrating films (OFDF) and cumulative dissolution profiles of sustained-release matrix tablets (SRMT). In the case of OFDF, we use principal component analysis (PCA) to reduce the dimensionality of the dataset, thereby improving the prediction performance and reducing the training time. In the case of SRMT, the Wasserstein generative adversarial network (WGAN), a neural network-based generative model, is used to overcome the limitation of performance improvement due to the lack of experimental data. To the best of our knowledge, this is the first work that utilizes WGAN for pharmaceutical formulation prediction. Experimental results show that the proposed methods have superior performance than the existing methods for all the performance metrics considered.
Collapse
Affiliation(s)
- Soyoung Yoo
- Department of Bigdata Engineering, Soonchunhyang University, Asan-si 31538, Korea;
| | - Junghyun Kim
- Department of Bigdata Engineering, Soonchunhyang University, Asan-si 31538, Korea;
- Department of Medical Sciences, Soonchunhyang University, Asan-si 31538, Korea;
| | - Guang J. Choi
- Department of Medical Sciences, Soonchunhyang University, Asan-si 31538, Korea;
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan-si 31538, Korea
| |
Collapse
|
47
|
Present and future challenges in therapeutic designing using computational approaches. COMPUTATIONAL APPROACHES FOR NOVEL THERAPEUTIC AND DIAGNOSTIC DESIGNING TO MITIGATE SARS-COV-2 INFECTION 2022. [PMCID: PMC9300749 DOI: 10.1016/b978-0-323-91172-6.00020-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Currently, various computational methods are being used for the purpose of therapeutic design. The advent of the Coronavirus disease-2019 (COVID-19) pandemic has created a lot of problems due to which the development of effective treatment options is urgently needed. Computational intelligence is used in the control, prevention, prediction, diagnosis, and treatment of the disease. Several important drug targets have been identified in severe acute respiratory syndrome-Coronavirus-2 using in silico methods. Computer-aided drug design includes a variety of theoretical and computational approaches that are part of modern drug discovery. Advances in machine learning methods and their applications speed up the drug discovery process. Exploration of nucleic acid-based therapeutics is playing an important role in healthcare also. But a lot of challenges have also been seen that complicate the therapeutic design. Therefore, investigation of challenges associated with therapeutic design is important, and the present chapter is aimed to cover various therapeutic design approaches and challenges associated with them. Moreover, the role of computational strategies in the exploration of potential therapeutics against COVID-19 has been investigated.
Collapse
|
48
|
Evaluation of the Effectiveness of Herbal Components Based on Their Regulatory Signature on Carcinogenic Cancer Cells. Cells 2021; 10:cells10113139. [PMID: 34831362 PMCID: PMC8621084 DOI: 10.3390/cells10113139] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/06/2021] [Accepted: 11/09/2021] [Indexed: 12/28/2022] Open
Abstract
Predicting cancer cells’ response to a plant-derived agent is critical for the drug discovery process. Recently transcriptomes advancements have provided an opportunity to identify regulatory signatures to predict drug activity. Here in this study, a combination of meta-analysis and machine learning models have been used to determine regulatory signatures focusing on differentially expressed transcription factors (TFs) of herbal components on cancer cells. In order to increase the size of the dataset, six datasets were combined in a meta-analysis from studies that had evaluated the gene expression in cancer cell lines before and after herbal extract treatments. Then, categorical feature analysis based on the machine learning methods was applied to examine transcription factors in order to find the best signature/pattern capable of discriminating between control and treated groups. It was found that this integrative approach could recognize the combination of TFs as predictive biomarkers. It was observed that the random forest (RF) model produced the best combination rules, including AIP/TFE3/VGLL4/ID1 and AIP/ZNF7/DXO with the highest modulating capacity. As the RF algorithm combines the output of many trees to set up an ultimate model, its predictive rules are more accurate and reproducible than other trees. The discovered regulatory signature suggests an effective procedure to figure out the efficacy of investigational herbal compounds on particular cells in the drug discovery process.
Collapse
|
49
|
Gaudelet T, Day B, Jamasb AR, Soman J, Regep C, Liu G, Hayter JBR, Vickers R, Roberts C, Tang J, Roblin D, Blundell TL, Bronstein MM, Taylor-King JP. Utilizing graph machine learning within drug discovery and development. Brief Bioinform 2021; 22:bbab159. [PMID: 34013350 PMCID: PMC8574649 DOI: 10.1093/bib/bbab159] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 12/15/2022] Open
Abstract
Graph machine learning (GML) is receiving growing interest within the pharmaceutical and biotechnology industries for its ability to model biomolecular structures, the functional relationships between them, and integrate multi-omic datasets - amongst other data types. Herein, we present a multidisciplinary academic-industrial review of the topic within the context of drug discovery and development. After introducing key terms and modelling approaches, we move chronologically through the drug development pipeline to identify and summarize work incorporating: target identification, design of small molecules and biologics, and drug repurposing. Whilst the field is still emerging, key milestones including repurposed drugs entering in vivo studies, suggest GML will become a modelling framework of choice within biomedical machine learning.
Collapse
Affiliation(s)
| | - Ben Day
- Relation Therapeutics, London, UK
- The Computer Laboratory, University of Cambridge, UK
| | - Arian R Jamasb
- Relation Therapeutics, London, UK
- The Computer Laboratory, University of Cambridge, UK
- Department of Biochemistry, University of Cambridge, UK
| | | | | | | | | | | | | | - Jian Tang
- Mila, the Quebec AI Institute, Canada
- HEC Montreal, Canada
| | - David Roblin
- Relation Therapeutics, London, UK
- Juvenescence, London, UK
- The Francis Crick Institute, London, UK
| | | | - Michael M Bronstein
- Relation Therapeutics, London, UK
- Department of Computing, Imperial College London, UK
- Twitter, UK
| | | |
Collapse
|
50
|
Golriz Khatami S, Mubeen S, Bharadhwaj VS, Kodamullil AT, Hofmann-Apitius M, Domingo-Fernández D. Using predictive machine learning models for drug response simulation by calibrating patient-specific pathway signatures. NPJ Syst Biol Appl 2021; 7:40. [PMID: 34707117 PMCID: PMC8551267 DOI: 10.1038/s41540-021-00199-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 09/21/2021] [Indexed: 11/21/2022] Open
Abstract
The utility of pathway signatures lies in their capability to determine whether a specific pathway or biological process is dysregulated in a given patient. These signatures have been widely used in machine learning (ML) methods for a variety of applications including precision medicine, drug repurposing, and drug discovery. In this work, we leverage highly predictive ML models for drug response simulation in individual patients by calibrating the pathway activity scores of disease samples. Using these ML models and an intuitive scoring algorithm to modify the signatures of patients, we evaluate whether a given sample that was formerly classified as diseased, could be predicted as normal following drug treatment simulation. We then use this technique as a proxy for the identification of potential drug candidates. Furthermore, we demonstrate the ability of our methodology to successfully identify approved and clinically investigated drugs for four different cancers, outperforming six comparable state-of-the-art methods. We also show how this approach can deconvolute a drugs' mechanism of action and propose combination therapies. Taken together, our methodology could be promising to support clinical decision-making in personalized medicine by simulating a drugs' effect on a given patient.
Collapse
Affiliation(s)
- Sepehr Golriz Khatami
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin, 53757, Germany.
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53115, Bonn, Germany.
| | - Sarah Mubeen
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin, 53757, Germany
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53115, Bonn, Germany
- Fraunhofer Center for Machine Learning, Sankt Augustin, Germany
| | - Vinay Srinivas Bharadhwaj
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin, 53757, Germany
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53115, Bonn, Germany
| | - Alpha Tom Kodamullil
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin, 53757, Germany
| | - Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin, 53757, Germany
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, 53115, Bonn, Germany
| | - Daniel Domingo-Fernández
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing, Sankt Augustin, 53757, Germany.
- Fraunhofer Center for Machine Learning, Sankt Augustin, Germany.
- Enveda Biosciences, Boulder, CO, 80301, USA.
| |
Collapse
|