1
|
Wawra-Hehenberger K, Pencelli V, Pardo JA, Tremmel L, Ataher Q, Waschbusch M. A visual safety risk evaluation tool in early clinical phases. Contemp Clin Trials 2025; 154:107953. [PMID: 40381907 DOI: 10.1016/j.cct.2025.107953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 05/09/2025] [Accepted: 05/10/2025] [Indexed: 05/20/2025]
Abstract
Dynamic evaluation of safety risks across all stages of clinical development, including early trial phases, has become the norm with the evolution of risk management in pharmacovigilance. The need for practical tools directly applicable to early clinical assessment prompted the authors to pilot the customization of a common risk matrix to support proactive safety risk evaluation based on multiple dimensions. Once defined for their research area, the tailored safety risk matrix was applied to visualize a series of known risk profiles and examples, along with projected risk mitigation effects. This two-fold visualization on one simple graph (risks positioning, and effect of risk mitigation of those positions) was found to be particularly relevant in multi-disciplinary safety discussions during early clinical development stages. The visual tool provides useful snapshots of projected safety risk profiles, facilitating communication with multiple stakeholders involved in decisions throughout early clinical development. With the prospect of a simple visual instrument to add to their existing risk management toolbox and processes, clinical teams are also provided with a basic blueprint to apply and tailor based on risk dimensions related to their own research area.
Collapse
|
2
|
de Siqueira Santos S, Yang H, Galeano A, Paccanaro A. Host centric drug repurposing for viral diseases. PLoS Comput Biol 2025; 21:e1012876. [PMID: 40173200 PMCID: PMC12052139 DOI: 10.1371/journal.pcbi.1012876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 05/05/2025] [Accepted: 02/14/2025] [Indexed: 04/04/2025] Open
Abstract
Computational approaches for drug repurposing for viral diseases have mainly focused on a small number of antivirals that directly target pathogens (virus centric therapies). In this work, we combine ideas from collaborative filtering and network medicine for making predictions on a much larger set of drugs that could be repurposed for host centric therapies, that are aimed at interfering with host cell factors required by a pathogen. Our idea is to create matrices quantifying the perturbation that drugs and viruses induce on human protein interaction networks. Then, we decompose these matrices to learn embeddings of drugs, viruses, and proteins in a low dimensional space. Predictions of host-centric antivirals are obtained by taking the dot product between the corresponding drug and virus representations. Our approach is general and can be applied systematically to any compound with known targets and any virus whose host proteins are known. We show that our predictions have high accuracy and that the embeddings contain meaningful biological information that may provide insights into the underlying biology of viral infections. Our approach can integrate different types of information, does not rely on known drug-virus associations and can be applied to new viral diseases and drugs.
Collapse
Affiliation(s)
| | - Haixuan Yang
- School of Mathematical & Statistical Sciences, University of Galway, Galway, Ireland
| | - Aldo Galeano
- Escola de Matemática Aplicada, Fundação Getúlio Vargas, Rio de Janeiro, Brazil
| | - Alberto Paccanaro
- Escola de Matemática Aplicada, Fundação Getúlio Vargas, Rio de Janeiro, Brazil
- Department of Computer Science, Centre for Systems and Synthetic Biology, Royal Holloway, University of London, Egham Hill, Egham, United Kingdom
| |
Collapse
|
3
|
Tanoli Z, Fernández-Torras A, Özcan UO, Kushnir A, Nader KM, Gadiya Y, Fiorenza L, Ianevski A, Vähä-Koskela M, Miihkinen M, Seemab U, Leinonen H, Seashore-Ludlow B, Tampere M, Kalman A, Ballante F, Benfenati E, Saunders G, Potdar S, Gómez García I, García-Serna R, Talarico C, Beccari AR, Schaal W, Polo A, Costantini S, Cabri E, Jacobs M, Saarela J, Budillon A, Spjuth O, Östling P, Xhaard H, Quintana J, Mestres J, Gribbon P, Ussi AE, Lo DC, de Kort M, Wennerberg K, Fratelli M, Carreras-Puigvert J, Aittokallio T. Computational drug repurposing: approaches, evaluation of in silico resources and case studies. Nat Rev Drug Discov 2025:10.1038/s41573-025-01164-x. [PMID: 40102635 DOI: 10.1038/s41573-025-01164-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2025] [Indexed: 03/20/2025]
Abstract
Repurposing of existing drugs for new indications has attracted substantial attention owing to its potential to accelerate drug development and reduce costs. Hundreds of computational resources such as databases and predictive platforms have been developed that can be applied for drug repurposing, making it challenging to select the right resource for a specific drug repurposing project. With the aim of helping to address this challenge, here we overview computational approaches to drug repurposing based on a comprehensive survey of available in silico resources using a purpose-built drug repurposing ontology that classifies the resources into hierarchical categories and provides application-specific information. We also present an expert evaluation of selected resources and three drug repurposing case studies implemented within the Horizon Europe REMEDi4ALL project to demonstrate the practical use of the resources. This comprehensive Review with expert evaluations and case studies provides guidelines and recommendations on the best use of various in silico resources for drug repurposing and establishes a basis for a sustainable and extendable drug repurposing web catalogue.
Collapse
Affiliation(s)
- Ziaurrehman Tanoli
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Drug Discovery and Chemical Biology (DDCB) Consortium, Biocenter Finland, University of Helsinki, Helsinki, Finland.
| | | | - Umut Onur Özcan
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Aleksandr Kushnir
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Kristen Michelle Nader
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Yojana Gadiya
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Frankfurt, Germany
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, Bonn, Germany
| | - Laura Fiorenza
- Dipartimento di Elettronica, Informazione e Bioingegneria (DEIB), Politecnico di Milano, Milan, Italy
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Aleksandr Ianevski
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Markus Vähä-Koskela
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mitro Miihkinen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Umair Seemab
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Henri Leinonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Brinton Seashore-Ludlow
- Science for Life Laboratory (SciLifeLab), Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Marianna Tampere
- Science for Life Laboratory (SciLifeLab), Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Adelinn Kalman
- Science for Life Laboratory (SciLifeLab), Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Flavio Ballante
- Chemical Biology Consortium Sweden (CBCS), SciLifeLab, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Emilio Benfenati
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Gary Saunders
- European Infrastructure for Translational Medicine (EATRIS ERIC), Amsterdam, The Netherlands
| | - Swapnil Potdar
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | | | | | | | | | - Wesley Schaal
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Andrea Polo
- Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Susan Costantini
- Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Enrico Cabri
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marc Jacobs
- Fraunhofer-Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, Germany
| | - Jani Saarela
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Alfredo Budillon
- Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Ola Spjuth
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Päivi Östling
- Science for Life Laboratory (SciLifeLab), Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Henri Xhaard
- Drug Discovery and Chemical Biology (DDCB) Consortium, Biocenter Finland, University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jordi Quintana
- Chemotargets SL, Parc Científic de Barcelona, Barcelona, Catalonia, Spain
| | - Jordi Mestres
- Chemotargets SL, Parc Científic de Barcelona, Barcelona, Catalonia, Spain
- Institut de Quimica Computacional i Catalisi, Facultat de Ciencies, Universitat de Girona, Girona, Catalonia, Spain
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Frankfurt, Germany
| | - Anton E Ussi
- European Infrastructure for Translational Medicine (EATRIS ERIC), Amsterdam, The Netherlands
| | - Donald C Lo
- European Infrastructure for Translational Medicine (EATRIS ERIC), Amsterdam, The Netherlands
| | - Martin de Kort
- European Infrastructure for Translational Medicine (EATRIS ERIC), Amsterdam, The Netherlands
| | - Krister Wennerberg
- Biotech Research & Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | | | - Jordi Carreras-Puigvert
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway.
- Oslo Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
4
|
Ma X, Wu T, Li G, Wang J, Jiang Y, Quan L, Lyu Q. DSE-HNGCN: Predicting the frequencies of drug-side effects based on heterogeneous networks with mining interactions between drugs and side effects. J Mol Biol 2025; 437:168916. [PMID: 39694183 DOI: 10.1016/j.jmb.2024.168916] [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: 07/08/2024] [Revised: 10/23/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024]
Abstract
Evaluating the frequencies of drug-side effects is crucial in drug development and risk-benefit analysis. While existing deep learning methods show promise, they have yet to explore using heterogeneous networks to simultaneously model the various relationship between drugs and side effects, highlighting areas for potential enhancement. In this study, we propose DSE-HNGCN, a novel method that leverages heterogeneous networks to simultaneously model the various relationships between drugs and side effects. By employing multi-layer graph convolutional networks, we aim to mine the interactions between drugs and side effects to predict the frequencies of drug-side effects. To address the over-smoothing problem in graph convolutional networks and capture diverse semantic information from different layers, we introduce a layer importance combination strategy. Additionally, we have developed an integrated prediction module that effectively utilizes drug and side effect features from different networks. Our experimental results, using benchmark datasets in a range of scenarios, show that our model outperforms existing methods in predicting the frequencies of drug-side effects. Comparative experiments and visual analysis highlight the substantial benefits of incorporating heterogeneous networks and other pertinent modules, thus improving the accuracy of DSE-HNGCN predictions. We also provide interpretability for DSE-HNGCN, indicating that the extracted features are potentially biologically significant. Case studies validate our model's capability to identify potential side effects of drugs, offering valuable insights for subsequent biological validation experiments.
Collapse
Affiliation(s)
- Xuhao Ma
- School of Computer Science and Technology, Soochow University, Ganjiang East Streat 333, 215006, Jiangsu, China
| | - Tingfang Wu
- School of Computer Science and Technology, Soochow University, Ganjiang East Streat 333, 215006, Jiangsu, China; Province Key Lab for Information Processing Technologies, Soochow University, Ganjiang East Streat 333, 215006, Jiangsu, China; Collaborative Innovation Center of Novel Software Technology and Industrialization, Organization, Nanjing 210000, Jiangsu, China.
| | - Geng Li
- School of Computer Science and Technology, Soochow University, Ganjiang East Streat 333, 215006, Jiangsu, China
| | - Junkai Wang
- School of Computer Science and Technology, Soochow University, Ganjiang East Streat 333, 215006, Jiangsu, China
| | - Yelu Jiang
- School of Computer Science and Technology, Soochow University, Ganjiang East Streat 333, 215006, Jiangsu, China
| | - Lijun Quan
- School of Computer Science and Technology, Soochow University, Ganjiang East Streat 333, 215006, Jiangsu, China; Province Key Lab for Information Processing Technologies, Soochow University, Ganjiang East Streat 333, 215006, Jiangsu, China; Collaborative Innovation Center of Novel Software Technology and Industrialization, Organization, Nanjing 210000, Jiangsu, China.
| | - Qiang Lyu
- School of Computer Science and Technology, Soochow University, Ganjiang East Streat 333, 215006, Jiangsu, China; Province Key Lab for Information Processing Technologies, Soochow University, Ganjiang East Streat 333, 215006, Jiangsu, China; Collaborative Innovation Center of Novel Software Technology and Industrialization, Organization, Nanjing 210000, Jiangsu, China.
| |
Collapse
|
5
|
Park S, Lee S, Pak M, Kim S. Dual Representation Learning for Predicting Drug-Side Effect Frequency Using Protein Target Information. IEEE J Biomed Health Inform 2025; 29:1817-1827. [PMID: 38241108 DOI: 10.1109/jbhi.2024.3350083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2024]
Abstract
Knowledge of unintended effects of drugs is critical in assessing the risk of treatment and in drug repurposing. Although numerous existing studies predict drug-side effect presence, only four of them predict the frequency of the side effects. Unfortunately, current prediction methods 1) do not utilize drug targets, 2) do not predict well for unseen drugs, and 3) do not use multiple heterogeneous drug features. We propose a novel deep learning-based drug-side effect frequency prediction model. Our model utilized heterogeneous features such as target protein information as well as molecular graph, fingerprints, and chemical similarity to create drug embeddings simultaneously. Furthermore, the model represents drugs and side effects into a common vector space, learning the dual representation vectors of drugs and side effects, respectively. We also extended the predictive power of our model to compensate for the drugs without clear target proteins using the Adaboost method. We achieved state-of-the-art performance over the existing methods in predicting side effect frequencies, especially for unseen drugs. Ablation studies show that our model effectively combines and utilizes heterogeneous features of drugs. Moreover, we observed that, when the target information given, drugs with explicit targets resulted in better prediction than the drugs without explicit targets.
Collapse
|
6
|
Yan D, Bao S, Zhang Z, Sun J, Zhou M. Leveraging pharmacovigilance data to predict population-scale toxicity profiles of checkpoint inhibitor immunotherapy. NATURE COMPUTATIONAL SCIENCE 2025; 5:207-220. [PMID: 39715829 DOI: 10.1038/s43588-024-00748-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/21/2024] [Indexed: 12/25/2024]
Abstract
Immune checkpoint inhibitor (ICI) therapies have made considerable advances in cancer immunotherapy, but the complex and diverse spectrum of ICI-induced toxicities poses substantial challenges to treatment outcomes and computational analysis. Here we introduce DySPred, a dynamic graph convolutional network-based deep learning framework, to map and predict the toxicity profiles of ICIs at the population level by leveraging large-scale real-world pharmacovigilance data. DySPred accurately predicts toxicity risks across diverse demographic cohorts and cancer types, demonstrating resilience in small-sample scenarios and revealing toxicity trends over time. Furthermore, DySPred consistently aligns the toxicity-safety profiles of small-molecule antineoplastic agents with their drug-induced transcriptional alterations. Our study provides a versatile methodology for population-level profiling of ICI-induced toxicities, enabling proactive toxicity monitoring and timely tailoring of treatment and intervention strategies in the advancement of cancer immunotherapy.
Collapse
Affiliation(s)
- Dongxue Yan
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Siqi Bao
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zicheng Zhang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jie Sun
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| | - Meng Zhou
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
7
|
Xuan P, Wu S, Cui H, Li P, Nakaguchi T, Zhang T. Interactive multi-hypergraph inferring and channel-enhanced and attribute-enhanced learning for drug-related side effect prediction. Comput Biol Med 2025; 184:109321. [PMID: 39522133 DOI: 10.1016/j.compbiomed.2024.109321] [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: 05/11/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Identifying the potential side effects for the interested drugs can help reduce harm to patients caused by drugs in clinical use and decrease the risk of drug development failure. Multiple functionally similar drugs often have multiple similar side effects, resulting in the closed relationships among these nodes. However, most of previous methods did not completely encode the features from the biological perspective to mine the complex associations between the drugs and side effects. A prediction model based on interactive multi-hypergraph inferring and channel-enhanced and attribute-enhanced learning, ICAL, was proposed to fuse the global correlations reflected by multiple hypergraphs and to learn the attributes of a pair of drug and side effect nodes enhanced by the channels and attributes. First, we designed a hypergraph architecture where a hyperedge reflects the complex correlations between a single drug (side effect) and all the drugs and side effects, and the entire hypergraph composed of the hyperedges reveals the global correlations of all the drugs and side effects. Two hypergraphs were established based on two types of drug similarities, and each hypergraph implies its specific complex relationships among multiple drugs and side effects. Second, we proposed an interactive hypergraph neural network to enable the learning of global correlation features of drugs and side effects from the two hypergraphs. It propagated the node features across multiple hypergraphs and encoded the context relationships within these hypergraphs. Besides, the attentions at the channel level and at the attribute level were proposed to integrate the semantic correlations among multiple channels and to encode the long-distance dependence within the attributes of a pair of drug and side effect. The experimental results based on cross-validation showed that our new model outperformed seven advanced prediction methods in terms of AUC, AUPR, and recall rates for the top-ranked candidates. The ablation studies showed the effectiveness of global correlation learning, node feature propagation across multiple hypergraphs, and channel and attribute enhanced pairwise attribute learning. The case studies on the candidate side effects related to five drugs further demonstrated ICAL's effective application in discovering the reliable candidates.
Collapse
Affiliation(s)
- Ping Xuan
- Department of Computer Science and Technology, Shantou University, Shantou, China; School of Cyberspace Security, Hainan University, Haikou, China
| | - Shien Wu
- Department of Computer Science and Technology, Shantou University, Shantou, China
| | - Hui Cui
- Department of Computer Science and Information Technology, La Trobe University, Australia; Australian Centre for AI in Medical Innovation, La Trobe University, Australia
| | - Peiru Li
- School of Computer Science and Technology, Heilongjiang University, Harbin, China
| | | | - Tiangang Zhang
- School of Cyberspace Security, Hainan University, Haikou, China.
| |
Collapse
|
8
|
Xu K, Wang M, Zou X, Liu J, Wei A, Chen J, Tang C. HSTrans: Homogeneous substructures transformer for predicting frequencies of drug-side effects. Neural Netw 2025; 181:106779. [PMID: 39488108 DOI: 10.1016/j.neunet.2024.106779] [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: 03/19/2024] [Revised: 08/29/2024] [Accepted: 10/01/2024] [Indexed: 11/04/2024]
Abstract
Identifying the frequencies of drug-side effects is crucial for assessing drug risk-benefit. However, accurately determining these frequencies remains challenging due to the limitations of time and scale in clinical randomized controlled trials. As a result, several computational methods have been proposed to address these issues. Nonetheless, two primary problems still persist. Firstly, most of these methods face challenges in generating accurate predictions for novel drugs, as they heavily depend on the interaction graph between drugs and side effects (SEs) within their modeling framework. Secondly, some previous methods often simply concatenate the features of drugs and SEs, which fails to effectively capture their underlying association. In this work, we present HSTrans, a novel approach that treats drugs and SEs as sets of substructures, leveraging a transformer encoder for unified substructure embedding and incorporating an interaction module for association capture. Specifically, HSTrans extracts drug substructures through a specialized algorithm and identifies effective substructures for each SE by employing an indicator that measures the importance of each substructure and SE. Additionally, HSTrans applies convolutional neural network (CNN) in the interaction module to capture complex relationships between drugs and SEs. Experimental results on datasets from Galeano et al.'s study demonstrate that the proposed method outperforms other state-of-the-art approaches. The demo codes for HSTrans are available at https://github.com/Dtdtxuky/HSTrans/tree/master.
Collapse
Affiliation(s)
- Kaiyi Xu
- School of Computer Science, China University of Geosciences, Wuhan 430074, China
| | - Minhui Wang
- Department of Pharmacy, Lianshui People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huai'an 223300, China
| | - Xin Zou
- School of Computer Science, China University of Geosciences, Wuhan 430074, China
| | - Jingjing Liu
- Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin 300222, China
| | - Ao Wei
- Department of Cardiology, Tianjin Chest Hospital, Tianjin 300222, China
| | - Jiajia Chen
- Department of Pharmacy, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an 223002, China.
| | - Chang Tang
- School of Computer Science, China University of Geosciences, Wuhan 430074, China.
| |
Collapse
|
9
|
Gao Y, Zhang X, Sun Z, Chandak P, Bu J, Wang H. Precision Adverse Drug Reactions Prediction with Heterogeneous Graph Neural Network. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 12:e2404671. [PMID: 39630592 PMCID: PMC11775569 DOI: 10.1002/advs.202404671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/11/2024] [Indexed: 12/07/2024]
Abstract
Accurate prediction of Adverse Drug Reactions (ADRs) at the patient level is essential for ensuring patient safety and optimizing healthcare outcomes. Traditional machine learning-based methods primarily focus on predicting potential ADRs for drugs, but they often fall short of capturing the complexity of individual demographics and the variations in ADRs experienced by different people. In this study, a novel framework called Precise Adverse Drug Reaction (PreciseADR) for patient-level ADR prediction is proposed. The approach effectively integrates relations between patients and ADRs, and harnesses the power of heterogeneous Graph Neural Networks (GNNs) to address the limitations of traditional methods. Specifically, a heterogeneous graph representation of patients is constructed, encompassing nodes that represent patients, diseases, drugs, and ADRs. By leveraging edges in the graph, crucial connections are captured such as a patient being affected by diseases, taking specific drugs, and experiencing ADRs. Next, a GNN-based model is utilized to learn latent representations of the patient nodes and facilitate the propagation of information throughout the graph structure. By employing patient embeddings that consider their diseases and drugs, potential ADRs can be accurately predicted. The PreciseADR is dedicated to effectively capturing both local and global dependencies within the heterogeneous graph, allowing for the identification of subtle patterns and interactions that play a significant role in ADRs. To evaluate the performance of the approach, extensive experiments are conducted on a large-scale real-world healthcare dataset with adverse reports from the FDA Adverse Event Reporting System (FAERS). Experimental results demonstrate that the PreciseADR achieves superior predictive performance in identifying patient-level ADRs, surpassing the strongest baseline by 3.2% in AUC score and by 4.9% in Hit@10.
Collapse
Affiliation(s)
- Yang Gao
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhou310009China
- College of Computer ScienceZhejiang UniversityHangzhou310058China
| | - Xiang Zhang
- Department of Computer ScienceThe University of North Carolina at CharlotteCharlotteNC28223‐0001USA
| | - Zhongquan Sun
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhou310009China
| | - Payal Chandak
- Harvard‐MIT Health Sciences and TechnologyCambridgeMA02139USA
| | - Jiajun Bu
- College of Computer ScienceZhejiang UniversityHangzhou310058China
| | - Haishuai Wang
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhou310009China
- College of Computer ScienceZhejiang UniversityHangzhou310058China
- Shanghai Artificial Intelligence LaboratoryShanghai200232China
| |
Collapse
|
10
|
Galeano D, Imrat, Haltom J, Andolino C, Yousey A, Zaksas V, Das S, Baylin SB, Wallace DC, Slack FJ, Enguita FJ, Wurtele ES, Teegarden D, Meller R, Cifuentes D, Beheshti A. sChemNET: a deep learning framework for predicting small molecules targeting microRNA function. Nat Commun 2024; 15:9149. [PMID: 39443444 PMCID: PMC11500171 DOI: 10.1038/s41467-024-49813-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 06/14/2024] [Indexed: 10/25/2024] Open
Abstract
MicroRNAs (miRNAs) have been implicated in human disorders, from cancers to infectious diseases. Targeting miRNAs or their target genes with small molecules offers opportunities to modulate dysregulated cellular processes linked to diseases. Yet, predicting small molecules associated with miRNAs remains challenging due to the small size of small molecule-miRNA datasets. Herein, we develop a generalized deep learning framework, sChemNET, for predicting small molecules affecting miRNA bioactivity based on chemical structure and sequence information. sChemNET overcomes the limitation of sparse chemical information by an objective function that allows the neural network to learn chemical space from a large body of chemical structures yet unknown to affect miRNAs. We experimentally validated small molecules predicted to act on miR-451 or its targets and tested their role in erythrocyte maturation during zebrafish embryogenesis. We also tested small molecules targeting the miR-181 network and other miRNAs using in-vitro and in-vivo experiments. We demonstrate that our machine-learning framework can predict bioactive small molecules targeting miRNAs or their targets in humans and other mammalian organisms.
Collapse
Affiliation(s)
- Diego Galeano
- Department of Electronics and Mechatronics Engineering, Facultad de Ingeniería, Universidad Nacional de Asunción - FIUNA, Luque, Paraguay.
- COVID-19 International Research Team, Medford, MA, USA.
| | - Imrat
- Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jeffrey Haltom
- COVID-19 International Research Team, Medford, MA, USA
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Chaylen Andolino
- Department of Nutrition Science, Purdue University, Indiana, USA
- Purdue Institute for Cancer Research, Purdue University, Indiana, USA
| | - Aliza Yousey
- COVID-19 International Research Team, Medford, MA, USA
- Neuroscience Institute, Department of Neurobiology/ Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Victoria Zaksas
- COVID-19 International Research Team, Medford, MA, USA
- Center for Translational Data Science, University of Chicago, Chicago, IL, USA
- Clever Research Lab, Springfield, IL, USA
| | - Saswati Das
- COVID-19 International Research Team, Medford, MA, USA
- Atal Bihari Vajpayee Institute of Medical Sciences and Dr Ram Manohar Lohia Hospital, New Delhi, India
| | - Stephen B Baylin
- COVID-19 International Research Team, Medford, MA, USA
- Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Van Andel Institute, Grand Rapids, MI, USA
| | - Douglas C Wallace
- COVID-19 International Research Team, Medford, MA, USA
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Frank J Slack
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Francisco J Enguita
- COVID-19 International Research Team, Medford, MA, USA
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Eve Syrkin Wurtele
- Bioinformatics and Computational Biology Program, Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, USA
| | - Dorothy Teegarden
- Department of Nutrition Science, Purdue University, Indiana, USA
- Purdue Institute for Cancer Research, Purdue University, Indiana, USA
| | - Robert Meller
- COVID-19 International Research Team, Medford, MA, USA
- Neuroscience Institute, Department of Neurobiology/ Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Daniel Cifuentes
- Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Virology, Immunology & Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Afshin Beheshti
- COVID-19 International Research Team, Medford, MA, USA
- Blue Marble Space Institute of Science, NASA Ames Research Center, Moffett Field, CA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- McGowan Institute for Regenerative Medicine - Center for Space Biomedicine, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
11
|
Baek B, Lee H. Crossfeat: a transformer-based cross-feature learning model for predicting drug side effect frequency. BMC Bioinformatics 2024; 25:324. [PMID: 39379821 PMCID: PMC11459996 DOI: 10.1186/s12859-024-05915-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/23/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Safe drug treatment requires an understanding of the potential side effects. Identifying the frequency of drug side effects can reduce the risks associated with drug use. However, existing computational methods for predicting drug side effect frequencies heavily depend on known drug side effect frequency information. Consequently, these methods face challenges when predicting the side effect frequencies of new drugs. Although a few methods can predict the side effect frequencies of new drugs, they exhibit unreliable performance owing to the exclusion of drug-side effect relationships. RESULTS This study proposed CrossFeat, a model based on convolutional neural network-transformer architecture with cross-feature learning that can predict the occurrence and frequency of drug side effects for new drugs, even in the absence of information regarding drug-side effect relationships. CrossFeat facilitates the concurrent learning of drugs and side effect information within its transformer architecture. This simultaneous exchange of information enables drugs to learn about their associated side effects, while side effects concurrently acquire information about the respective drugs. Such bidirectional learning allows for the comprehensive integration of drug and side effect knowledge. Our five-fold cross-validation experiments demonstrated that CrossFeat outperforms existing studies in predicting side effect frequencies for new drugs without prior knowledge. CONCLUSIONS Our model offers a promising approach for predicting the drug side effect frequencies, particularly for new drugs where prior information is limited. CrossFeat's superior performance in cross-validation experiments, along with evidence from case studies and ablation experiments, highlights its effectiveness.
Collapse
Affiliation(s)
- Bin Baek
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
| | - Hyunju Lee
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea.
- AI Graduate School, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea.
| |
Collapse
|
12
|
Song Z, Chen G, Chen CYC. AI empowering traditional Chinese medicine? Chem Sci 2024; 15:d4sc04107k. [PMID: 39355231 PMCID: PMC11440359 DOI: 10.1039/d4sc04107k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/22/2024] [Indexed: 10/03/2024] Open
Abstract
For centuries, Traditional Chinese Medicine (TCM) has been a prominent treatment method in China, incorporating acupuncture, herbal remedies, massage, and dietary therapy to promote holistic health and healing. TCM has played a major role in drug discovery, with over 60% of small-molecule drugs approved by the FDA from 1981 to 2019 being derived from natural products. However, TCM modernization faces challenges such as data standardization and the complexity of TCM formulations. The establishment of comprehensive TCM databases has significantly improved the efficiency and accuracy of TCM research, enabling easier access to information on TCM ingredients and encouraging interdisciplinary collaborations. These databases have revolutionized TCM research, facilitating advancements in TCM modernization and patient care. In addition, advancements in AI algorithms and database data quality have accelerated progress in AI for TCM. The application of AI in TCM encompasses a wide range of areas, including herbal screening and new drug discovery, diagnostic and treatment principles, pharmacological mechanisms, network pharmacology, and the incorporation of innovative AI technologies. AI also has the potential to enable personalized medicine by identifying patterns and correlations in patient data, leading to more accurate diagnoses and tailored treatments. The potential benefits of AI for TCM are vast and diverse, promising continued progress and innovation in the field.
Collapse
Affiliation(s)
- Zhilin Song
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Shenzhen Guangdong 518055 China
- AI for Science (AI4S)-Preferred Program, School of Electronic and Computer Engineering, Peking University Shenzhen Graduate School Shenzhen Guangdong 518055 China
| | - Guanxing Chen
- Artificial Intelligence Medical Research Center, School of Intelligent Systems Engineering, Shenzhen Campus of Sun Yat-sen University Shenzhen Guangdong 518107 China
| | - Calvin Yu-Chian Chen
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Shenzhen Guangdong 518055 China
- AI for Science (AI4S)-Preferred Program, School of Electronic and Computer Engineering, Peking University Shenzhen Graduate School Shenzhen Guangdong 518055 China
- Department of Medical Research, China Medical University Hospital Taichung 40447 Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University Taichung 41354 Taiwan
- Guangdong L-Med Biotechnology Co., Ltd Meizhou Guangdong 514699 China
| |
Collapse
|
13
|
Funari A, Fiscon G, Paci P. Network medicine and systems pharmacology approaches to predicting adverse drug effects. Br J Pharmacol 2024. [PMID: 39262113 DOI: 10.1111/bph.17330] [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: 03/18/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 09/13/2024] Open
Abstract
Identifying and understanding the relationships between drug intake and adverse effects that can occur due to inadvertent molecular interactions between drugs and targets is a difficult task, especially considering the numerous variables that can influence the onset of such events. The ability to predict these side effects in advance would help physicians develop strategies to avoid or counteract them. In this article, we review the main computational methods for predicting side effects caused by drug molecules, highlighting their performance, limitations and application cases. Furthermore, we provide an overall view of resources, such as databases and tools, useful for building side effect prediction analyses.
Collapse
Affiliation(s)
- Alessio Funari
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
- Institute for Systems Analysis and Computer Science "Antonio Ruberti", National Research Council, Rome, Italy
| | - Giulia Fiscon
- Institute for Systems Analysis and Computer Science "Antonio Ruberti", National Research Council, Rome, Italy
- Department of Computer, Control and Management Engineering "Antonio Ruberti", Sapienza University of Rome, Rome, Italy
| | - Paola Paci
- Institute for Systems Analysis and Computer Science "Antonio Ruberti", National Research Council, Rome, Italy
- Department of Computer, Control and Management Engineering "Antonio Ruberti", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
14
|
Cao A, Zhang L, Bu Y, Sun D. Machine Learning Prediction of On/Off Target-driven Clinical Adverse Events. Pharm Res 2024; 41:1649-1658. [PMID: 39095534 DOI: 10.1007/s11095-024-03742-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 07/06/2024] [Indexed: 08/04/2024]
Abstract
OBJECTIVE Currently, 90% of clinical drug development fails, where 30% of these failures are due to clinical toxicity. The current extensive animal toxicity studies are not predictive of clinical adverse events (AEs) at clinical doses, while current computation models only consider very few factors with limited success in clinical toxicity prediction. We aimed to address these issues by developing a machine learning (ML) model to directly predict clinical AEs. METHODS Using a dataset with 759 FDA-approved drugs with known AEs, we first adapted the ConPLex ML model to predict IC50 values of these FDA-approved drugs against their on-target and off-target binding among 477 protein targets. Subsequently, we constructed a new ML model to predict clinical AEs using IC50 values of 759 drugs' primary on-target and off-target effects along with tissue-specific protein expression profiles. RESULTS The adapted ConPLex model predicted drug-target interactions for both on- and off-target effects, as shown by co-localization of the 6 small molecule kinase inhibitors with their respective kinases. The coupled ML models demonstrated good predictive capability of clinical AEs, with accuracy over 75%. CONCLUSIONS Our approach provides a new insight into the mechanistic understanding of in vivo drug toxicity in relationship with drug on-/off-target interactions. The coupled ML models, once validated with larger datasets, may offer advantages to directly predict clinical AEs using in vitro/ex vivo and preclinical data, which will help to reduce drug development failure due to clinical toxicity.
Collapse
Affiliation(s)
- Albert Cao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, United States
- Centennial High School, Ellicott City, MD, 21042, United States
| | - Luchen Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Yingzi Bu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, United States
- Michigan Institute for Computational Discovery & Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, United States.
- Duxin Sun, 1600 Huron Parkway, North Campus Research Complex, Building 520, Ann Arbor, MI, 48109, United States.
| |
Collapse
|
15
|
Kenakin T. Know your molecule: pharmacological characterization of drug candidates to enhance efficacy and reduce late-stage attrition. Nat Rev Drug Discov 2024; 23:626-644. [PMID: 38890494 DOI: 10.1038/s41573-024-00958-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2024] [Indexed: 06/20/2024]
Abstract
Despite advances in chemical, computational and biological sciences, the rate of attrition of drug candidates in clinical development is still high. A key point in the small-molecule discovery process that could provide opportunities to help address this challenge is the pharmacological characterization of hit and lead compounds, culminating in the selection of a drug candidate. Deeper characterization is increasingly important, because the 'quality' of drug efficacy, at least for G protein-coupled receptors (GPCRs), is now understood to be much more than activation of commonly evaluated pathways such as cAMP signalling, with many more 'efficacies' of ligands that could be harnessed therapeutically. Such characterization is being enabled by novel assays to characterize the complex behaviour of GPCRs, such as biased signalling and allosteric modulation, as well as advances in structural biology, such as cryo-electron microscopy. This article discusses key factors in the assessments of the pharmacology of hit and lead compounds in the context of GPCRs as a target class, highlighting opportunities to identify drug candidates with the potential to address limitations of current therapies and to improve the probability of them succeeding in clinical development.
Collapse
Affiliation(s)
- Terry Kenakin
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
16
|
Zhang H, Zhou Y, Zhang Z, Sun H, Pan Z, Mou M, Zhang W, Ye Q, Hou T, Li H, Hsieh CY, Zhu F. Large Language Model-Based Natural Language Encoding Could Be All You Need for Drug Biomedical Association Prediction. Anal Chem 2024. [PMID: 39011990 DOI: 10.1021/acs.analchem.4c01793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Analyzing drug-related interactions in the field of biomedicine has been a critical aspect of drug discovery and development. While various artificial intelligence (AI)-based tools have been proposed to analyze drug biomedical associations (DBAs), their feature encoding did not adequately account for crucial biomedical functions and semantic concepts, thereby still hindering their progress. Since the advent of ChatGPT by OpenAI in 2022, large language models (LLMs) have demonstrated rapid growth and significant success across various applications. Herein, LEDAP was introduced, which uniquely leveraged LLM-based biotext feature encoding for predicting drug-disease associations, drug-drug interactions, and drug-side effect associations. Benefiting from the large-scale knowledgebase pre-training, LLMs had great potential in drug development analysis owing to their holistic understanding of natural language and human topics. LEDAP illustrated its notable competitiveness in comparison with other popular DBA analysis tools. Specifically, even in simple conjunction with classical machine learning methods, LLM-based feature representations consistently enabled satisfactory performance across diverse DBA tasks like binary classification, multiclass classification, and regression. Our findings underpinned the considerable potential of LLMs in drug development research, indicating a catalyst for further progress in related fields.
Collapse
Affiliation(s)
- Hanyu Zhang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Yuan Zhou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhichao Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Huaicheng Sun
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ziqi Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wei Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qing Ye
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tingjun Hou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Honglin Li
- Innovation Center for AI and Drug Discovery, East China Normal University, Shanghai 200062, China
| | - Chang-Yu Hsieh
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhu
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
17
|
Hauben M, Rafi M, Abdelaziz I, Hassanzadeh O. Knowledge Graphs in Pharmacovigilance: A Scoping Review. Clin Ther 2024; 46:544-554. [PMID: 38981792 DOI: 10.1016/j.clinthera.2024.06.003] [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: 12/12/2023] [Revised: 05/08/2024] [Accepted: 06/05/2024] [Indexed: 07/11/2024]
Abstract
PURPOSE To critically assess the role and added value of knowledge graphs in pharmacovigilance, focusing on their ability to predict adverse drug reactions. METHODS A systematic scoping review was conducted in which detailed information, including objectives, technology, data sources, methodology, and performance metrics, were extracted from a set of peer-reviewed publications reporting the use of knowledge graphs to support pharmacovigilance signal detection. FINDINGS The review, which included 47 peer-reviewed articles, found knowledge graphs were utilized for detecting/predicting single-drug adverse reactions and drug-drug interactions, with variable reported performance and sparse comparisons to legacy methods. IMPLICATIONS Research to date suggests that knowledge graphs have the potential to augment predictive signal detection in pharmacovigilance, but further research using more reliable reference sets of adverse drug reactions and comparison with legacy pharmacovigilance methods are needed to more clearly define best practices and to establish their place in holistic pharmacovigilance systems.
Collapse
Affiliation(s)
- Manfred Hauben
- Department of Family and Community Medicine, New York Medical College, Valhalla, New York; Truliant Consulting, Baltimore, Maryland
| | - Mazin Rafi
- Department of Statistics, Rutgers University, Piscataway, New Jersey.
| | | | | |
Collapse
|
18
|
Yang J, Hu Z, Zhang L, Peng B. Predicting Drugs Suspected of Causing Adverse Drug Reactions Using Graph Features and Attention Mechanisms. Pharmaceuticals (Basel) 2024; 17:822. [PMID: 39065673 PMCID: PMC11279999 DOI: 10.3390/ph17070822] [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: 05/27/2024] [Revised: 06/12/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Adverse drug reactions (ADRs) refer to an unintended harmful reaction that occurs after the administration of a medication for therapeutic purposes, which is unrelated to the intended pharmacological action of the drug. In the United States, ADRs account for 6% of all hospital admissions annually. The cost of ADR-related illnesses in 2016 was estimated at USD 528.4 billion. Increasing the awareness of ADRs is an effective measure to prevent them. Assessing suspected drugs in adverse events helps to enhance the awareness of ADRs. METHODS In this study, a suspect drug assisted judgment model (SDAJM) is designed to identify suspected drugs in adverse events. This framework utilizes the graph isomorphism network (GIN) and an attention mechanism to extract features based on patients' demographic information, drug information, and ADR information. RESULTS By comparing it with other models, the results of various tests show that this model performs well in predicting the suspected drugs in adverse reaction events. ADR signal detection was conducted on a group of cardiovascular system drugs, and case analyses were performed on two classic drugs, Mexiletine and Captopril, as well as on two classic antithyroid drugs. The results indicate that the model can accomplish the task of predicting drug ADRs. Validation using benchmark datasets from ten drug discovery domains shows that the model is applicable to classification tasks on the Tox21 and SIDER datasets. CONCLUSIONS This study applies deep learning methods to construct the SDAJM model for three purposes: (1) identifying drugs suspected to cause adverse drug events (ADEs), (2) predicting the ADRs of drugs, and (3) other drug discovery tasks. The results indicate that this method can offer new directions for research in the field of ADRs.
Collapse
Affiliation(s)
| | | | | | - Bin Peng
- College of Public Health, Chongqing Medical University, Chongqing 401331, China; (J.Y.); (Z.H.); (L.Z.)
| |
Collapse
|
19
|
Bai H, Lu S, Zhang T, Cui H, Nakaguchi T, Xuan P. Graph reasoning method enhanced by relational transformers and knowledge distillation for drug-related side effect prediction. iScience 2024; 27:109571. [PMID: 38799562 PMCID: PMC11126883 DOI: 10.1016/j.isci.2024.109571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/29/2023] [Accepted: 03/22/2024] [Indexed: 05/29/2024] Open
Abstract
Identifying the side effects related to drugs is beneficial for reducing the risk of drug development failure and saving the drug development cost. We proposed a graph reasoning method, RKDSP, to fuse the semantics of multiple connection relationships, the local knowledge within each meta-path, the global knowledge among multiple meta-paths, and the attributes of the drug and side effect node pairs. We constructed drug-side effect heterogeneous graphs consisting of the drugs, side effects, and their similarity and association connections. Multiple relational transformers were established to learn node features from diverse meta-path semantic perspectives. A knowledge distillation module was constructed to learn local and global knowledge of multiple meta-paths. Finally, an adaptive convolutional neural network-based strategy was presented to adaptively encode the attributes of each drug-side effect node pair. The experimental results demonstrated that RKDSP outperforms the compared state-of-the-art prediction approaches.
Collapse
Affiliation(s)
- Honglei Bai
- School of Computer Science and Technology, Heilongjiang University, Harbin, China
| | - Siyuan Lu
- School of Computer Science and Technology, Heilongjiang University, Harbin, China
| | - Tiangang Zhang
- School of Computer Science and Technology, Heilongjiang University, Harbin, China
- School of Mathematical Science, Heilongjiang University, Harbin, China
| | - Hui Cui
- Department of Computer Science and Information Technology, La Trobe University, Melbourne, VIC, Australia
| | - Toshiya Nakaguchi
- Center for Frontier Medical Engineering, Chiba University, Chiba, Japan
| | - Ping Xuan
- Department of Computer Science and Technology, Shantou University, Shantou, China
| |
Collapse
|
20
|
Toni E, Ayatollahi H, Abbaszadeh R, Fotuhi Siahpirani A. Machine Learning Techniques for Predicting Drug-Related Side Effects: A Scoping Review. Pharmaceuticals (Basel) 2024; 17:795. [PMID: 38931462 PMCID: PMC11206653 DOI: 10.3390/ph17060795] [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: 04/13/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Drug safety relies on advanced methods for timely and accurate prediction of side effects. To tackle this requirement, this scoping review examines machine-learning approaches for predicting drug-related side effects with a particular focus on chemical, biological, and phenotypical features. METHODS This was a scoping review in which a comprehensive search was conducted in various databases from 1 January 2013 to 31 December 2023. RESULTS The results showed the widespread use of Random Forest, k-nearest neighbor, and support vector machine algorithms. Ensemble methods, particularly random forest, emphasized the significance of integrating chemical and biological features in predicting drug-related side effects. CONCLUSIONS This review article emphasized the significance of considering a variety of features, datasets, and machine learning algorithms for predicting drug-related side effects. Ensemble methods and Random Forest showed the best performance and combining chemical and biological features improved prediction. The results suggested that machine learning techniques have some potential to improve drug development and trials. Future work should focus on specific feature types, selection techniques, and graph-based methods for even better prediction.
Collapse
Affiliation(s)
- Esmaeel Toni
- Medical Informatics, Student Research Committee, Iran University of Medical Sciences, Tehran, Iran 14496-14535;
| | - Haleh Ayatollahi
- Medical Informatics, Health Management and Economics Research Center, Health Management Research Institute, Iran University of Medical Sciences, Tehran, Iran 1996-713883
| | - Reza Abbaszadeh
- Pediatric Cardiology, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran 19956-14331;
| | - Alireza Fotuhi Siahpirani
- Systems Biology and Bioinformatics, Department of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran 14176-14411;
| |
Collapse
|
21
|
Liu W, Zhang J, Qiao G, Bian J, Dong B, Li Y. HMMF: a hybrid multi-modal fusion framework for predicting drug side effect frequencies. BMC Bioinformatics 2024; 25:196. [PMID: 38769492 PMCID: PMC11555943 DOI: 10.1186/s12859-024-05806-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/08/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND The identification of drug side effects plays a critical role in drug repositioning and drug screening. While clinical experiments yield accurate and reliable information about drug-related side effects, they are costly and time-consuming. Computational models have emerged as a promising alternative to predict the frequency of drug-side effects. However, earlier research has primarily centered on extracting and utilizing representations of drugs, like molecular structure or interaction graphs, often neglecting the inherent biomedical semantics of drugs and side effects. RESULTS To address the previously mentioned issue, we introduce a hybrid multi-modal fusion framework (HMMF) for predicting drug side effect frequencies. Considering the wealth of biological and chemical semantic information related to drugs and side effects, incorporating multi-modal information offers additional, complementary semantics. HMMF utilizes various encoders to understand molecular structures, biomedical textual representations, and attribute similarities of both drugs and side effects. It then models drug-side effect interactions using both coarse and fine-grained fusion strategies, effectively integrating these multi-modal features. CONCLUSIONS HMMF exhibits the ability to successfully detect previously unrecognized potential side effects, demonstrating superior performance over existing state-of-the-art methods across various evaluation metrics, including root mean squared error and area under receiver operating characteristic curve, and shows remarkable performance in cold-start scenarios.
Collapse
Affiliation(s)
- Wuyong Liu
- College of Computer and Control Engineering, Northeast Forestry University, Harbin, 150006, China
| | - Jingyu Zhang
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Guanyu Qiao
- Computer Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Jilong Bian
- College of Computer and Control Engineering, Northeast Forestry University, Harbin, 150006, China
| | - Benzhi Dong
- College of Computer and Control Engineering, Northeast Forestry University, Harbin, 150006, China
| | - Yang Li
- College of Computer and Control Engineering, Northeast Forestry University, Harbin, 150006, China.
| |
Collapse
|
22
|
Habib M, Lalagkas PN, Melamed RD. Mapping drug biology to disease genetics to discover drug impacts on the human phenome. BIOINFORMATICS ADVANCES 2024; 4:vbae038. [PMID: 38736684 PMCID: PMC11087821 DOI: 10.1093/bioadv/vbae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/18/2024] [Accepted: 03/07/2024] [Indexed: 05/14/2024]
Abstract
Motivation Medications can have unexpected effects on disease, including not only harmful drug side effects, but also beneficial drug repurposing. These effects on disease may result from hidden influences of drugs on disease gene networks. Then, discovering how biological effects of drugs relate to disease biology can both provide insight into the mechanism of latent drug effects, and can help predict new effects. Results Here, we develop Draphnet, a model that integrates molecular data on 429 drugs and gene associations of nearly 200 common phenotypes to learn a network that explains drug effects on disease in terms of these molecular signals. We present evidence that our method can both predict drug effects, and can provide insight into the biology of unexpected drug effects on disease. Using Draphnet to map a drug's known molecular effects to downstream effects on the disease genome, we put forward disease genes impacted by drugs, and we suggest a new grouping of drugs based on shared effects on the disease genome. Our approach has multiple applications, including predicting drug uses and learning drug biology, with implications for personalized medicine. Availability and implementation Code to reproduce the analysis is available at https://github.com/RDMelamed/drug-phenome.
Collapse
Affiliation(s)
- Mamoon Habib
- Department of Computer Science, University of Massachusetts Lowell, Lowell, MA 01854, United States
| | | | - Rachel D Melamed
- Department of Biological Science, University of Massachusetts Lowell, Lowell, MA 01854, United States
| |
Collapse
|
23
|
Ali MT, Johnson M, Irwin T, Henry S, Sugeng L, Kansal S, Allison TG, Bremer ML, Jones VR, Martineau MD, Wong C, Marecki G, Stebbins J, Michelena HI, McCully RB, Svatikova A, Padang R, Scott CG, Kanuga MJ, Arsanjani R, Pellikka PA, Kane GC, Thaden JJ. Incidence of Severe Adverse Drug Reactions to Ultrasound Enhancement Agents in a Contemporary Echocardiography Practice. J Am Soc Echocardiogr 2024; 37:276-284.e3. [PMID: 37879379 DOI: 10.1016/j.echo.2023.10.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/02/2023] [Accepted: 10/12/2023] [Indexed: 10/27/2023]
Abstract
OBJECTIVES Prior data indicate a very rare risk of serious adverse drug reaction (ADR) to ultrasound enhancement agents (UEAs). We sought to evaluate the frequency of ADR to UEA administration in contemporary practice. METHODS We retrospectively reviewed 4 US health systems to characterize the frequency and severity of ADR to UEA. Adverse drug reactions were considered severe when cardiopulmonary involvement was present and critical when there was loss of consciousness, loss of pulse, or ST-segment elevation. Rates of isolated back pain and headache were derived from the Mayo Clinic Rochester stress echocardiography database where systematic prospective reporting of ADR was performed. RESULTS Among 26,539 Definity and 11,579 Lumason administrations in the Mayo Clinic Rochester stress echocardiography database, isolated back pain or headache was more frequent with Definity (0.49% vs 0.04%, P < .0001) but less common with Definity infusion versus bolus (0.08% vs 0.53%, P = .007). Among all sites there were 201,834 Definity and 84,943 Lumason administrations. Severe and critical ADR were more frequent with Lumason than with Definity (0.0848% vs 0.0114% and 0.0330% vs 0.0010%, respectively; P < .001 for each). Among the 3 health systems with >2,000 Lumason administrations, the frequency of severe ADR with Lumason ranged from 0.0755% to 0.1093% and the frequency of critical ADR ranged from 0.0293% to 0.0525%. Severe ADR rates with Definity were stable over time but increased in more recent years with Lumason (P = .02). Patients with an ADR to Lumason since the beginning of 2021 were more likely to have received a COVID-19 vaccination compared with matched controls (88% vs 75%; P = .05) and more likely to have received Moderna than Pfizer-Biotech (71% vs 26%, P < .001). CONCLUSION Severe and critical ADR, while rare, were more frequent with Lumason, and the frequency has increased in more recent years. Additional work is needed to better understand factors, including associations with recently developed mRNA vaccines, which may be contributing to the increased rates of ADR to UEA since 2021.
Collapse
Affiliation(s)
- Mays T Ali
- Division of Cardiovascular Ultrasound, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Mark Johnson
- Division of Cardiovascular Ultrasound, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Timothy Irwin
- University of South Dakota, Yankton Medical Clinic, Yankton, South Dakota
| | - Sonia Henry
- Department of Cardiology, Northwell Health, Manhasset, New York
| | - Lissa Sugeng
- Department of Cardiology, Northwell Health, Manhasset, New York
| | - Sarita Kansal
- WellStar Center for Cardiovascular Medicine, WellStar Health System, Atlanta, Georgia
| | - Thomas G Allison
- Division of Cardiovascular Ultrasound, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota; Department of Pediatric and Adolescent Medicine, Division of Pediatric Cardiology, Mayo Clinic, Rochester, Minnesota
| | - Merri L Bremer
- Division of Cardiovascular Ultrasound, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Victoria R Jones
- Division of Cardiovascular Ultrasound, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Michael D Martineau
- Division of Cardiovascular Ultrasound, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Connie Wong
- Department of Cardiology, Northwell Health, Manhasset, New York
| | - Gregory Marecki
- Department of Cardiology, Northwell Health, Manhasset, New York
| | - Julie Stebbins
- WellStar Center for Cardiovascular Medicine, WellStar Health System, Atlanta, Georgia
| | - Hector I Michelena
- Division of Cardiovascular Ultrasound, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Robert B McCully
- Division of Cardiovascular Ultrasound, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Anna Svatikova
- Division of Cardiovascular Ultrasound, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Ratnasari Padang
- Division of Cardiovascular Ultrasound, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Christopher G Scott
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Mansi J Kanuga
- Division of Allergic Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Reza Arsanjani
- Division of Cardiac Imaging and Stress Testing, Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona
| | - Patricia A Pellikka
- Division of Cardiovascular Ultrasound, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Garvan C Kane
- Division of Cardiovascular Ultrasound, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Jeremy J Thaden
- Division of Cardiovascular Ultrasound, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
24
|
Seal S, Spjuth O, Hosseini-Gerami L, García-Ortegón M, Singh S, Bender A, Carpenter AE. Insights into Drug Cardiotoxicity from Biological and Chemical Data: The First Public Classifiers for FDA Drug-Induced Cardiotoxicity Rank. J Chem Inf Model 2024; 64:1172-1186. [PMID: 38300851 PMCID: PMC10900289 DOI: 10.1021/acs.jcim.3c01834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 02/03/2024]
Abstract
Drug-induced cardiotoxicity (DICT) is a major concern in drug development, accounting for 10-14% of postmarket withdrawals. In this study, we explored the capabilities of chemical and biological data to predict cardiotoxicity, using the recently released DICTrank data set from the United States FDA. We found that such data, including protein targets, especially those related to ion channels (e.g., hERG), physicochemical properties (e.g., electrotopological state), and peak concentration in plasma offer strong predictive ability for DICT. Compounds annotated with mechanisms of action such as cyclooxygenase inhibition could distinguish between most-concern and no-concern DICT. Cell Painting features for ER stress discerned most-concern cardiotoxic from nontoxic compounds. Models based on physicochemical properties provided substantial predictive accuracy (AUCPR = 0.93). With the availability of omics data in the future, using biological data promises enhanced predictability and deeper mechanistic insights, paving the way for safer drug development. All models from this study are available at https://broad.io/DICTrank_Predictor.
Collapse
Affiliation(s)
- Srijit Seal
- Imaging
Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Ola Spjuth
- Department
of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Box
591, SE-75124 Uppsala, Sweden
| | - Layla Hosseini-Gerami
- Ignota
Labs, The Bradfield Centre, Cambridge Science Park, County Hall, Westminster Bridge Road, Cambridge CB4 0GA, U.K.
| | - Miguel García-Ortegón
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Shantanu Singh
- Imaging
Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Andreas Bender
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Anne E. Carpenter
- Imaging
Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
25
|
Cheng F, Wang F, Tang J, Zhou Y, Fu Z, Zhang P, Haines JL, Leverenz JB, Gan L, Hu J, Rosen-Zvi M, Pieper AA, Cummings J. Artificial intelligence and open science in discovery of disease-modifying medicines for Alzheimer's disease. Cell Rep Med 2024; 5:101379. [PMID: 38382465 PMCID: PMC10897520 DOI: 10.1016/j.xcrm.2023.101379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 08/15/2023] [Accepted: 12/19/2023] [Indexed: 02/23/2024]
Abstract
The high failure rate of clinical trials in Alzheimer's disease (AD) and AD-related dementia (ADRD) is due to a lack of understanding of the pathophysiology of disease, and this deficit may be addressed by applying artificial intelligence (AI) to "big data" to rapidly and effectively expand therapeutic development efforts. Recent accelerations in computing power and availability of big data, including electronic health records and multi-omics profiles, have converged to provide opportunities for scientific discovery and treatment development. Here, we review the potential utility of applying AI approaches to big data for discovery of disease-modifying medicines for AD/ADRD. We illustrate how AI tools can be applied to the AD/ADRD drug development pipeline through collaborative efforts among neurologists, gerontologists, geneticists, pharmacologists, medicinal chemists, and computational scientists. AI and open data science expedite drug discovery and development of disease-modifying therapeutics for AD/ADRD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA.
| | - Fei Wang
- Department of Population Health Sciences, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Jian Tang
- Mila-Quebec Institute for Learning Algorithms and CIFAR AI Research Chair, HEC Montreal, Montréal, QC H3T 2A7, Canada
| | - Yadi Zhou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Zhimin Fu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Pengyue Zhang
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN 46037, USA
| | - Jonathan L Haines
- Cleveland Institute for Computational Biology, and Department of Population & Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jianying Hu
- IBM Research, Yorktown Heights, New York, NY 10598, USA
| | - Michal Rosen-Zvi
- AI for Accelerated Healthcare and Life Sciences Discovery, IBM Research Labs, Haifa 3498825, Israel; Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9190500, Israel
| | - Andrew A Pieper
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH 44106, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland OH 44106, USA; Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA; Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, UNLV, Las Vegas, NV 89154, USA
| |
Collapse
|
26
|
Li J, Zhu T, Jiang Y, Zhang Q, Zu Y, Shen X. Microfluidic printed 3D bioactive scaffolds for postoperative treatment of gastric cancer. Mater Today Bio 2024; 24:100911. [PMID: 38188649 PMCID: PMC10770549 DOI: 10.1016/j.mtbio.2023.100911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/04/2023] [Accepted: 12/09/2023] [Indexed: 01/09/2024] Open
Abstract
Tumor recurrence and tissue regeneration are two major challenges in the postoperative treatment of cancer. Current research hotspots are focusing on developing novel scaffold materials that can simultaneously suppress tumor recurrence and promote tissue repair. Here, we propose a microfluidic 3D-printed methacrylate fish gelatin (F-GelMA@BBR) scaffold loaded with berberine (BBR) for the postoperative treatment of gastric cancer. The F-GelMA@BBR scaffold displayed a significant killing effect on gastric cancer MKN-45 cells in vitro and demonstrated excellent anti-recurrence efficiency in gastric cancer postoperative models. In vitro experiments have shown that F-GelMA@BBR exhibits significant cytotoxicity on gastric cancer cells while maintaining the cell viability of normal cells. The results of in vivo experiments show that F-GelMA@BBR can significantly suppress the tumor volume to 49.7 % of the control group. In addition, the scaffold has an ordered porous structure and good biocompatibility, which could support the attachment and proliferation of normal cells to promote tissue repair at the tumor resection site. These features indicated that such scaffold material is a promising candidate for postoperative tumor treatment in the practical application.
Collapse
Affiliation(s)
- Jiante Li
- Department of Anorectal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Tianru Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yiwei Jiang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Qingfei Zhang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
- The Key Laboratory of Pediatric Hematology and Oncology Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yan Zu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Xian Shen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| |
Collapse
|
27
|
Traipop S, Jesadabundit W, Khamcharoen W, Pholsiri T, Naorungroj S, Jampasa S, Chailapakul O. Nanomaterial-based Electrochemical Sensors for Multiplex Medicinal Applications. Curr Top Med Chem 2024; 24:986-1009. [PMID: 38584544 DOI: 10.2174/0115680266304711240327072348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/07/2024] [Accepted: 03/15/2024] [Indexed: 04/09/2024]
Abstract
This review explores the advancements in nanomaterial-based electrochemical sensors for the multiplex detection of medicinal compounds. The growing demand for efficient and selective detection methods in the pharmaceutical field has prompted significant research into the development of electrochemical sensors employing nanomaterials. These materials, defined as functional materials with at least one dimension between 1 and 100 nanometers, encompass metal nanoparticles, polymers, carbon-based nanocomposites, and nano-bioprobes. These sensors are characterized by their enhanced sensitivity and selectivity, playing a crucial role in simultaneous detection and offering a comprehensive analysis of multiple medicinal complexes within a single sample. The review comprehensively examines the design, fabrication, and application of nanomaterial- based electrochemical sensors, focusing on their ability to achieve multiplex detection of various medicinal substances. Insights into the strategies and nanomaterials employed for enhancing sensor performance are discussed. Additionally, the review explores the challenges and future perspectives of this evolving field, highlighting the potential impact of nanomaterial-based electrochemical sensors on the advancement of medicinal detection technologies.
Collapse
Affiliation(s)
- Surinya Traipop
- Electrochemistry and Optical Spectroscopy Center of Excellence (EOSCE), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Whitchuta Jesadabundit
- Electrochemistry and Optical Spectroscopy Center of Excellence (EOSCE), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Wisarut Khamcharoen
- Electrochemistry and Optical Spectroscopy Center of Excellence (EOSCE), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence on Petrochemical and Materials Technology (PETROMAT), Thailand
| | - Tavechai Pholsiri
- Electrochemistry and Optical Spectroscopy Center of Excellence (EOSCE), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Sarida Naorungroj
- Electrochemistry and Optical Spectroscopy Center of Excellence (EOSCE), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Sakda Jampasa
- Electrochemistry and Optical Spectroscopy Center of Excellence (EOSCE), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Orawon Chailapakul
- Electrochemistry and Optical Spectroscopy Center of Excellence (EOSCE), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| |
Collapse
|
28
|
Shtar G, Solomon A, Mazuz E, Rokach L, Shapira B. A simplified similarity-based approach for drug-drug interaction prediction. PLoS One 2023; 18:e0293629. [PMID: 37943768 PMCID: PMC10635435 DOI: 10.1371/journal.pone.0293629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/17/2023] [Indexed: 11/12/2023] Open
Abstract
Drug-drug interactions (DDIs) are a critical component of drug safety surveillance. Laboratory studies aimed at detecting DDIs are typically difficult, expensive, and time-consuming; therefore, developing in-silico methods is critical. Machine learning-based approaches for DDI prediction have been developed; however, in many cases, their ability to achieve high accuracy relies on data only available towards the end of the molecule lifecycle. Here, we propose a simple yet effective similarity-based method for preclinical DDI prediction where only the chemical structure is available. We test the model on new, unseen drugs. To focus on the preclinical problem setting, we conducted a retrospective analysis and tested the models on drugs that were added to a later version of the DrugBank database. We extend an existing method, adjacency matrix factorization with propagation (AMFP), to support unseen molecules by applying a new lookup mechanism to the drugs' chemical structure, lookup adjacency matrix factorization with propagation (LAMFP). We show that using an ensemble of different similarity measures improves the results. We also demonstrate that Chemprop, a message-passing neural network, can be used for DDI prediction. In computational experiments, LAMFP results in high accuracy, with an area under the receiver operating characteristic curve of 0.82 for interactions involving a new drug and an existing drug and for interactions involving only existing drugs. Moreover, LAMFP outperforms state-of-the-art, complex graph neural network DDI prediction methods.
Collapse
Affiliation(s)
- Guy Shtar
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Information Systems, University of Haifa, Haifa, Israel
| | - Adir Solomon
- Department of Information Systems, University of Haifa, Haifa, Israel
| | - Eyal Mazuz
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Lior Rokach
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Bracha Shapira
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
29
|
Ugbe FA, Shallangwa GA, Uzairu A, Abdulkadir I, Edache EI, Al-Megrin WAI, Al-Shouli ST, Wang Y, Abdalla M. Cheminformatics-based discovery of new organoselenium compounds with potential for the treatment of cutaneous and visceral leishmaniasis. J Biomol Struct Dyn 2023; 42:13830-13853. [PMID: 37937770 DOI: 10.1080/07391102.2023.2279269] [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: 07/25/2023] [Accepted: 10/30/2023] [Indexed: 11/09/2023]
Abstract
Leishmaniasis affects more than 12 million humans globally and a further 1 billion people are at risk in leishmaniasis endemic areas. The lack of a vaccine for leishmaniasis coupled with the limitations of existing anti-leishmanial therapies prompted this study. Cheminformatic techniques are widely used in screening large libraries of compounds, studying protein-ligand interactions, analysing pharmacokinetic properties, and designing new drug molecules with great speed, accuracy, and precision. This study was undertaken to evaluate the anti-leishmanial potential of some organoselenium compounds by quantitative structure-activity relationship (QSAR) modeling, molecular docking, pharmacokinetic analysis, and molecular dynamic (MD) simulation. The built QSAR model was validated (R2train = 0.8646, R2test = 0.8864, Q2 = 0.5773) and the predicted inhibitory activity (pIC50) values of the newly designed compounds were higher than that of the template (Compound 6). The new analogues (6a, 6b, and 6c) showed good binding interactions with the target protein (Pyridoxal kinase, PdxK) while also presenting excellent drug-likeness and pharmacokinetic profiles. The results of density functional theory, MD simulation, and molecular mechanics generalized Born surface area (MM/GBSA) analyses suggest the favourability and stability of protein-ligand interactions of the new analogues with PdxK, comparing favourably well with the reference drug (Pentamidine). Conclusively, the newly designed compounds could be synthesized and tested experimentally as potential anti-leishmanial drug molecules.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Fabian Audu Ugbe
- Department of Chemistry, Faculty of Physical Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Gideon Adamu Shallangwa
- Department of Chemistry, Faculty of Physical Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Adamu Uzairu
- Department of Chemistry, Faculty of Physical Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Ibrahim Abdulkadir
- Department of Chemistry, Faculty of Physical Sciences, Ahmadu Bello University, Zaria, Nigeria
| | | | - Wafa Abdullah I Al-Megrin
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman. University, Riyadh, Saudi Arabia
| | - Samia T Al-Shouli
- Immunology Unit, Pathology Department, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ying Wang
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Children's Health and Disease, Jinan, Shandong, China
| | - Mohnad Abdalla
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Children's Health and Disease, Jinan, Shandong, China
| |
Collapse
|
30
|
Seal S, Spjuth O, Hosseini-Gerami L, García-Ortegón M, Singh S, Bender A, Carpenter AE. Insights into Drug Cardiotoxicity from Biological and Chemical Data: The First Public Classifiers for FDA DICTrank. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.15.562398. [PMID: 37905146 PMCID: PMC10614794 DOI: 10.1101/2023.10.15.562398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Drug-induced cardiotoxicity (DICT) is a major concern in drug development, accounting for 10-14% of postmarket withdrawals. In this study, we explored the capabilities of various chemical and biological data to predict cardiotoxicity, using the recently released Drug-Induced Cardiotoxicity Rank (DICTrank) dataset from the United States FDA. We analyzed a diverse set of data sources, including physicochemical properties, annotated mechanisms of action (MOA), Cell Painting, Gene Expression, and more, to identify indications of cardiotoxicity. We found that such data, including protein targets, especially those related to ion channels (such as hERG), physicochemical properties (such as electrotopological state) as well as peak concentration in plasma offer strong predictive ability as well as valuable insights into DICT. We also found compounds annotated with particular mechanisms of action, such as cyclooxygenase inhibition, could distinguish between most-concern and no-concern DICT compounds. Cell Painting features related to ER stress discern the most-concern cardiotoxic compounds from non-toxic compounds. While models based on physicochemical properties currently provide substantial predictive accuracy (AUCPR = 0.93), this study also underscores the potential benefits of incorporating more comprehensive biological data in future DICT predictive models. With the availability of - omics data in the future, using biological data promises enhanced predictability and delivers deeper mechanistic insights, paving the way for safer therapeutic drug development. All models and data used in this study are publicly released at https://broad.io/DICTrank_Predictor.
Collapse
Affiliation(s)
- Srijit Seal
- Imaging Platform, Broad Institute of MIT and Harvard, US
| | - Ola Spjuth
- Department of Pharmaceutical Biosciences, Uppsala University, Sweden
| | | | | | - Shantanu Singh
- Imaging Platform, Broad Institute of MIT and Harvard, US
| | | | | |
Collapse
|
31
|
Xuan P, Li P, Cui H, Wang M, Nakaguchi T, Zhang T. Learning Multi-Types of Neighbor Node Attributes and Semantics by Heterogeneous Graph Transformer and Multi-View Attention for Drug-Related Side-Effect Prediction. Molecules 2023; 28:6544. [PMID: 37764319 PMCID: PMC10537290 DOI: 10.3390/molecules28186544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Since side-effects of drugs are one of the primary reasons for their failure in clinical trials, predicting their side-effects can help reduce drug development costs. We proposed a method based on heterogeneous graph transformer and capsule networks for side-effect-drug-association prediction (TCSD). The method encodes and integrates attributes from multiple types of neighbor nodes, connection semantics, and multi-view pairwise information. In each drug-side-effect heterogeneous graph, a target node has two types of neighbor nodes, the drug nodes and the side-effect ones. We proposed a new heterogeneous graph transformer-based context representation learning module. The module is able to encode specific topology and the contextual relations among multiple kinds of nodes. There are similarity and association connections between the target node and its various types of neighbor nodes, and these connections imply semantic diversity. Therefore, we designed a new strategy to measure the importance of a neighboring node to the target node and incorporate different semantics of the connections between the target node and its multi-type neighbors. Furthermore, we designed attentions at the neighbor node type level and at the graph level, respectively, to obtain enhanced informative neighbor node features and multi-graph features. Finally, a pairwise multi-view feature learning module based on capsule networks was built to learn the pairwise attributes from the heterogeneous graphs. Our prediction model was evaluated using a public dataset, and the cross-validation results showed it achieved superior performance to several state-of-the-art methods. Ablation experiments undertaken demonstrated the effectiveness of heterogeneous graph transformer-based context encoding, the position enhanced pairwise attribute learning, and the neighborhood node category-level attention. Case studies on five drugs further showed TCSD's ability in retrieving potential drug-related side-effect candidates, and TCSD inferred the candidate side-effects for 708 drugs.
Collapse
Affiliation(s)
- Ping Xuan
- School of Computer Science and Technology, Heilongjiang University, Harbin 130407, China
- Department of Computer Science, School of Engineering, Shantou University, Shantou 515000, China
| | - Peiru Li
- School of Computer Science and Technology, Heilongjiang University, Harbin 130407, China
| | - Hui Cui
- Department of Computer Science and Information Technology, La Trobe University, Melbourne 3086, Australia
| | - Meng Wang
- School of Computer Science and Technology, Heilongjiang University, Harbin 130407, China
| | - Toshiya Nakaguchi
- Center for Frontier Medical Engineering, Chiba University, Chiba 263-8522, Japan
| | - Tiangang Zhang
- School of Computer Science and Technology, Heilongjiang University, Harbin 130407, China
- School of Mathematical Science, Heilongjiang University, Harbin 130407, China
| |
Collapse
|
32
|
Xuan P, Xu K, Cui H, Nakaguchi T, Zhang T. Graph generative and adversarial strategy-enhanced node feature learning and self-calibrated pairwise attribute encoding for prediction of drug-related side effects. Front Pharmacol 2023; 14:1257842. [PMID: 37731739 PMCID: PMC10507253 DOI: 10.3389/fphar.2023.1257842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 08/17/2023] [Indexed: 09/22/2023] Open
Abstract
Background: Inferring drug-related side effects is beneficial for reducing drug development cost and time. Current computational prediction methods have concentrated on graph reasoning over heterogeneous graphs comprising the drug and side effect nodes. However, the various topologies and node attributes within multiple drug-side effect heterogeneous graphs have not been completely exploited. Methods: We proposed a new drug-side effect association prediction method, GGSC, to deeply integrate the diverse topologies and attributes from multiple heterogeneous graphs and the self-calibration attributes of each drug-side effect node pair. First, we created two heterogeneous graphs comprising the drug and side effect nodes and their related similarity and association connections. Since each heterogeneous graph has its specific topology and node attributes, a node feature learning strategy was designed and the learning for each graph was enhanced from a graph generative and adversarial perspective. We constructed a generator based on a graph convolutional autoencoder to encode the topological structure and node attributes from the whole heterogeneous graph and then generate the node features embedding the graph topology. A discriminator based on multilayer perceptron was designed to distinguish the generated topological features from the original ones. We also designed representation-level attention to discriminate the contributions of topological representations from multiple heterogeneous graphs and adaptively fused them. Finally, we constructed a self-calibration module based on convolutional neural networks to guide pairwise attribute learning through the features of the small latent space. Results: The comparison experiment results showed that GGSC had higher prediction performance than several state-of-the-art prediction methods. The ablation experiments demonstrated the effectiveness of topological enhancement learning, representation-level attention, and self-calibrated pairwise attribute learning. In addition, case studies over five drugs demonstrated GGSC's ability in discovering the potential drug-related side effect candidates. Conclusion: We proposed a drug-side effect association prediction method, and the method is beneficial for screening the reliable association candidates for the biologists to discover the actual associations.
Collapse
Affiliation(s)
- Ping Xuan
- Department of Computer Science, School of Engineering, Shantou University, Shantou, China
| | - Kai Xu
- School of Computer Science and Technology, Heilongjiang University, Harbin, China
| | - Hui Cui
- Department of Computer Science and Information Technology, La Trobe University, Melbourne, VI, Australia
| | - Toshiya Nakaguchi
- Center for Frontier Medical Engineering, Chiba University, Chiba, Japan
| | - Tiangang Zhang
- School of Computer Science and Technology, Heilongjiang University, Harbin, China
- School of Mathematical Science, Heilongjiang University, Harbin, China
| |
Collapse
|
33
|
Wang L, Sun C, Xu X, Li J, Zhang W. A neighborhood-regularization method leveraging multiview data for predicting the frequency of drug-side effects. Bioinformatics 2023; 39:btad532. [PMID: 37647657 PMCID: PMC10491955 DOI: 10.1093/bioinformatics/btad532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/01/2023] Open
Abstract
MOTIVATION A critical issue in drug benefit-risk assessment is to determine the frequency of side effects, which is performed by randomized controlled trails. Computationally predicted frequencies of drug side effects can be used to effectively guide the randomized controlled trails. However, it is more challenging to predict drug side effect frequencies, and thus only a few studies cope with this problem. RESULTS In this work, we propose a neighborhood-regularization method (NRFSE) that leverages multiview data on drugs and side effects to predict the frequency of side effects. First, we adopt a class-weighted non-negative matrix factorization to decompose the drug-side effect frequency matrix, in which Gaussian likelihood is used to model unknown drug-side effect pairs. Second, we design a multiview neighborhood regularization to integrate three drug attributes and two side effect attributes, respectively, which makes most similar drugs and most similar side effects have similar latent signatures. The regularization can adaptively determine the weights of different attributes. We conduct extensive experiments on one benchmark dataset, and NRFSE improves the prediction performance compared with five state-of-the-art approaches. Independent test set of post-marketing side effects further validate the effectiveness of NRFSE. AVAILABILITY AND IMPLEMENTATION Source code and datasets are available at https://github.com/linwang1982/NRFSE or https://codeocean.com/capsule/4741497/tree/v1.
Collapse
Affiliation(s)
- Lin Wang
- College of Artificial Intelligence, Tianjin University of Science and Technology, No. 9, 13th Street, Tianjin Economic-Technological Development Area, Tianjin 300457, China
| | - Chenhao Sun
- College of Artificial Intelligence, Tianjin University of Science and Technology, No. 9, 13th Street, Tianjin Economic-Technological Development Area, Tianjin 300457, China
| | - Xianyu Xu
- College of Artificial Intelligence, Tianjin University of Science and Technology, No. 9, 13th Street, Tianjin Economic-Technological Development Area, Tianjin 300457, China
| | - Jia Li
- College of Artificial Intelligence, Tianjin University of Science and Technology, No. 9, 13th Street, Tianjin Economic-Technological Development Area, Tianjin 300457, China
| | - Wenjuan Zhang
- College of General Education, Tianjin Foreign Studies University, No. 117, Machang Road, Hexi District, Tianjin 300204, China
| |
Collapse
|
34
|
Zhao H, Ni P, Zhao Q, Liang X, Ai D, Erhardt S, Wang J, Li Y, Wang J. Identifying the serious clinical outcomes of adverse reactions to drugs by a multi-task deep learning framework. Commun Biol 2023; 6:870. [PMID: 37620651 PMCID: PMC10449791 DOI: 10.1038/s42003-023-05243-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
Adverse Drug Reactions (ADRs) have a direct impact on human health. As continuous pharmacovigilance and drug monitoring prove to be costly and time-consuming, computational methods have emerged as promising alternatives. However, most existing computational methods primarily focus on predicting whether or not the drug is associated with an adverse reaction and do not consider the core issue of drug benefit-risk assessment-whether the treatment outcome is serious when adverse drug reactions occur. To this end, we categorize serious clinical outcomes caused by adverse reactions to drugs into seven distinct classes and present a deep learning framework, so-called GCAP, for predicting the seriousness of clinical outcomes of adverse reactions to drugs. GCAP has two tasks: one is to predict whether adverse reactions to drugs cause serious clinical outcomes, and the other is to infer the corresponding classes of serious clinical outcomes. Experimental results demonstrate that our method is a powerful and robust framework with high extendibility. GCAP can serve as a useful tool to successfully address the challenge of predicting the seriousness of clinical outcomes stemming from adverse reactions to drugs.
Collapse
Affiliation(s)
- Haochen Zhao
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha, 410083, China
- Xiangjiang Laboratory, Changsha, 410205, China
| | - Peng Ni
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha, 410083, China
| | - Qichang Zhao
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha, 410083, China
| | - Xiao Liang
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha, 410083, China
| | - Di Ai
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Shannon Erhardt
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yaohang Li
- Department of Computer Science, Old Dominion University, Norfolk, VA, 23529-0001, USA
| | - Jianxin Wang
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China.
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha, 410083, China.
- Xiangjiang Laboratory, Changsha, 410205, China.
| |
Collapse
|
35
|
Huang T, Lin KH, Machado-Vieira R, Soares JC, Jiang X, Kim Y. Explainable drug side effect prediction via biologically informed graph neural network. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.26.23290615. [PMID: 37333107 PMCID: PMC10275013 DOI: 10.1101/2023.05.26.23290615] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Early detection of potential side effects (SE) is a critical and challenging task for drug discovery and patient care. In-vitro or in-vivo approach to detect potential SEs is not scalable for many drug candidates during the preclinical stage. Recent advances in explainable machine learning may facilitate detecting potential SEs of new drugs before market release and elucidating the critical mechanism of biological actions. Here, we leverage multi-modal interactions among molecules to develop a biologically informed graph-based SE prediction model, called HHAN-DSI. HHAN-DSI predicted frequent and even uncommon SEs of the unseen drug with higher or comparable accuracy against benchmark methods. When applying HHAN-DSI to the central nervous system, the organs with the largest number of SEs, the model revealed diverse psychiatric medications' previously unknown but probable SEs, together with the potential mechanisms of actions through a network of genes, biological functions, drugs, and SEs.
Collapse
Affiliation(s)
- Tongtong Huang
- School of Biomedical Informatics, UTHealth, Houston, TX, United States
| | - Ko-Hong Lin
- School of Biomedical Informatics, UTHealth, Houston, TX, United States
| | - Rodrigo Machado-Vieira
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, UTHealth, Houston, TX, United States
| | - Jair C Soares
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, UTHealth, Houston, TX, United States
| | - Xiaoqian Jiang
- School of Biomedical Informatics, UTHealth, Houston, TX, United States
| | - Yejin Kim
- School of Biomedical Informatics, UTHealth, Houston, TX, United States
| |
Collapse
|
36
|
Menéndez-Velázquez A, García-Delgado AB. A Novel Photopharmacological Tool: Dual-Step Luminescence for Biological Tissue Penetration of Light and the Selective Activation of Photodrugs. Int J Mol Sci 2023; 24:ijms24119404. [PMID: 37298355 DOI: 10.3390/ijms24119404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Conventional pharmacology lacks spatial and temporal selectivity in terms of drug action. This leads to unwanted side effects, such as damage to healthy cells, as well as other less obvious effects, such as environmental toxicity and the acquisition of resistance to drugs, especially antibiotics, by pathogenic microorganisms. Photopharmacology, based on the selective activation of drugs by light, can contribute to alleviating this serious problem. However, many of these photodrugs are activated by light in the UV-visible spectral range, which does not propagate through biological tissues. In this article, to overcome this problem, we propose a dual-spectral conversion technique, which simultaneously makes use of up-conversion (using rare earth elements) and down-shifting (using organic materials) techniques in order to modify the spectrum of light. Near-infrared light (980 nm), which penetrates tissue fairly well, can provide a "remote control" for drug activation. Once near-IR light is inside the body, it is up-converted to the UV-visible spectral range. Subsequently, this radiation is down-shifted in order to accurately adjust to the excitation wavelengths of light which can selectively activate hypothetical and specific photodrugs. In summary, this article presents, for the first time, a "dual tunable light source" which can penetrate into the human body and deliver light of specific wavelengths; thus, it can overcome one of the main limitations of photopharmacology. It opens up promising possibilities for the moving of photodrugs from the laboratory to the clinic.
Collapse
|
37
|
Asif M, Aqil F, Alasmary FA, almalki AS, Khan AR, Nasibullah M. Lewis base-catalyzed synthesis of highly functionalized spirooxindole-pyranopyrazoles and their in vitro anticancer studies. Med Chem Res 2023. [DOI: 10.1007/s00044-023-03053-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
|
38
|
Das P, Mazumder DH. An extensive survey on the use of supervised machine learning techniques in the past two decades for prediction of drug side effects. Artif Intell Rev 2023; 56:1-28. [PMID: 36819660 PMCID: PMC9930028 DOI: 10.1007/s10462-023-10413-7] [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] [Accepted: 02/01/2023] [Indexed: 02/19/2023]
Abstract
Approved drugs for sale must be effective and safe, implying that the drug's advantages outweigh its known harmful side effects. Side effects (SE) of drugs are one of the common reasons for drug failure that may halt the whole drug discovery pipeline. The side effects might vary from minor concerns like a runny nose to potentially life-threatening issues like liver damage, heart attack, and death. Therefore, predicting the side effects of the drug is vital in drug development, discovery, and design. Supervised machine learning-based side effects prediction task has recently received much attention since it reduces time, chemical waste, design complexity, risk of failure, and cost. The advancement of supervised learning approaches for predicting side effects have emerged as essential computational tools. Supervised machine learning technique provides early information on drug side effects to develop an effective drug based on drug properties. Still, there are several challenges to predicting drug side effects. Thus, a near-exhaustive survey is carried out in this paper on the use of supervised machine learning approaches employed in drug side effects prediction tasks in the past two decades. In addition, this paper also summarized the drug descriptor required for the side effects prediction task, commonly utilized drug properties sources, computational models, and their performances. Finally, the research gap, open problems, and challenges for the further supervised learning-based side effects prediction task have been discussed.
Collapse
Affiliation(s)
- Pranab Das
- Department of Computer Science and Engineering, National Institute of Technology Nagaland, Chumukedima, Dimapur, Nagaland 797103 India
| | - Dilwar Hussain Mazumder
- Department of Computer Science and Engineering, National Institute of Technology Nagaland, Chumukedima, Dimapur, Nagaland 797103 India
| |
Collapse
|
39
|
Azuma I, Mizuno T, Kusuhara H. NRBdMF: A Recommendation Algorithm for Predicting Drug Effects Considering Directionality. J Chem Inf Model 2023; 63:474-483. [PMID: 36635231 DOI: 10.1021/acs.jcim.2c01210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Predicting the novel effects of drugs based on information about approved drugs can be regarded as a recommendation system. Matrix factorization is one of the most used recommendation systems, and various algorithms have been devised for it. A literature survey and summary of existing algorithms for predicting drug effects demonstrated that most such methods, including neighborhood regularized logistic matrix factorization, which was the best performer in benchmark tests, used a binary matrix that considers only the presence or absence of interactions. However, drug effects are known to have two opposite aspects, such as side effects and therapeutic effects. In the present study, we proposed using neighborhood regularized bidirectional matrix factorization (NRBdMF) to predict drug effects by incorporating bidirectionality, which is a characteristic property of drug effects. We used this proposed method for predicting side effects using a matrix that considered the bidirectionality of drug effects, in which known side effects were assigned a positive (+1) label and known treatment effects were assigned a negative (-1) label. The NRBdMF model, which utilizes drug bidirectional information, achieved enrichment of side effects at the top and indications at the bottom of the prediction list. This first attempt to consider the bidirectional nature of drug effects using NRBdMF showed that it reduced false positives and produced a highly interpretable output.
Collapse
Affiliation(s)
- Iori Azuma
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo113-0033, Japan
| | - Tadahaya Mizuno
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo113-0033, Japan
| | - Hiroyuki Kusuhara
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo113-0033, Japan
| |
Collapse
|
40
|
Galeano D, Paccanaro A. Machine learning prediction of side effects for drugs in clinical trials. CELL REPORTS METHODS 2022; 2:100358. [PMID: 36590692 PMCID: PMC9795366 DOI: 10.1016/j.crmeth.2022.100358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 09/08/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022]
Abstract
Early and accurate detection of side effects is critical for the clinical success of drugs under development. Here, we aim to predict unknown side effects for drugs with a small number of side effects identified in randomized controlled clinical trials. Our machine learning framework, the geometric self-expressive model (GSEM), learns globally optimal self-representations for drugs and side effects from pharmacological graph networks. We show the usefulness of the GSEM on 505 therapeutically diverse drugs and 904 side effects from multiple human physiological systems. Here, we also show a data integration strategy that could be adopted to improve the ability of side effect prediction models to identify unknown side effects that might only appear after the drug enters the market.
Collapse
Affiliation(s)
- Diego Galeano
- Department of Electronics and Mechatronics Engineering, Facultad de Ingeniería, Universidad Nacional de Asunción, San Lorenzo, Paraguay
| | - Alberto Paccanaro
- School of Applied Mathematics, Fundação Getulio Vargas, Rio de Janeiro, Brazil
- Department of Computer Science, Centre for Systems and Synthetic Biology, Royal Holloway, University of London, Egham Hill, Egham, UK
| |
Collapse
|
41
|
Qian Y, Ding Y, Zou Q, Guo F. Identification of drug-side effect association via restricted Boltzmann machines with penalized term. Brief Bioinform 2022; 23:6762741. [PMID: 36259601 DOI: 10.1093/bib/bbac458] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/09/2022] [Accepted: 09/25/2022] [Indexed: 12/14/2022] Open
Abstract
In the entire life cycle of drug development, the side effect is one of the major failure factors. Severe side effects of drugs that go undetected until the post-marketing stage leads to around two million patient morbidities every year in the United States. Therefore, there is an urgent need for a method to predict side effects of approved drugs and new drugs. Following this need, we present a new predictor for finding side effects of drugs. Firstly, multiple similarity matrices are constructed based on the association profile feature and drug chemical structure information. Secondly, these similarity matrices are integrated by Centered Kernel Alignment-based Multiple Kernel Learning algorithm. Then, Weighted K nearest known neighbors is utilized to complement the adjacency matrix. Next, we construct Restricted Boltzmann machines (RBM) in drug space and side effect space, respectively, and apply a penalized maximum likelihood approach to train model. At last, the average decision rule was adopted to integrate predictions from RBMs. Comparison results and case studies demonstrate, with four benchmark datasets, that our method can give a more accurate and reliable prediction result.
Collapse
Affiliation(s)
- Yuqing Qian
- School of Electronic and Information Engineering, Suzhou University of Science and Technology, Suzhou 215009, PR China
| | - Yijie Ding
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324000, PR China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610054, PR China
| | - Fei Guo
- School of Computer Science and Engineering, Central South University, Changsha 410083, PR China
| |
Collapse
|
42
|
Alpay BA, Gosink M, Aguiar D. Evaluating molecular fingerprint-based models of drug side effects against a statistical control. Drug Discov Today 2022; 27:103364. [PMID: 36115633 DOI: 10.1016/j.drudis.2022.103364] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/24/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022]
Abstract
There are many machine learning models that use molecular fingerprints of drugs to predict side effects. Characterizing their skill is necessary for understanding their usefulness in pharmaceutical development. Here, we analyze a statistical control of side effect prediction skill, develop a pipeline for benchmarking models, and evaluate how well existing models predict side effects identified in pharmaceutical documentation. We demonstrate that molecular fingerprints are useful for ranking drugs by their likelihood to cause a given side effect. However, the predictions for one or more drugs overall benefit only marginally from molecular fingerprints when ranking the likelihoods of many possible side effects, and display at most modest overall skill at identifying the side effects that do and do not occur.
Collapse
Affiliation(s)
- Berk A Alpay
- Systems, Synthetic, and Quantitative Biology Program, Harvard University, Cambridge, MA 02138, USA.
| | | | - Derek Aguiar
- Department of Computer Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
43
|
Flores LE, Elgart JF, Abraham AG, Garrote GL, Torrieri R, Cepeda A, Cardelle-Cobas A, Gagliardino JJ. Changes in lifestyle behaviors during COVID-19 isolation in Argentina: A cross-sectional study. Nutr Health 2022:2601060221127115. [PMID: 36221976 PMCID: PMC9554566 DOI: 10.1177/02601060221127115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Our aim was to identify changes in population habits induced by COVID-19 confinement in Argentina. METHODS An internet-based cross-sectional survey was conducted among adults in Argentina on December 2020, requesting possible changes occurring during the COVID-19 outbreak. It included 26 questions regarding general information (age, gender, location), eating habits, desire/anxiety for food or to eat between meals, weight gain, physical activity, and hours of sleep. We ran a descriptive statistical analysis of changes in habits and lifestyle during the confinement, followed by a logistic regression analysis to explore the relation between these changes and weight gain. Results: Out of 1536 survey participants, 57.1% were female, aged 38.8 ± 13.1 years. Data showed that during the outbreak, people experienced significant changes in food intake, physical activity, nutritional supplement consumption, anxiety, and sleeping disorders. These changes in behavior resulted in an elevated percentage of people (39.7%) that gained weight (average 4.8 ± 2.8 kg). Weight gain was associated with more food consumption (OR: 9.398), increased snacking between meals (OR: 1.536), anxiety about food (OR: 3.180), less practice of physical activity (OR: 0.586) and less consumption of nutritional supplements (OR: 0.762). Conclusions: COVID-19 outbreak was associated with unhealthy lifestyle changes and body weight increase. These adverse side effects could be prevented by active promotion of nutritional advice and physical activity, implementing virtual activities associated with regular mass promotion campaigns.
Collapse
Affiliation(s)
- Luis E. Flores
- Centro de Endocrinología Experimental y Aplicada (UNLP-CONICET-CeAs CICPBA), Facultad de Ciencias Médicas UNLP, La Plata, Argentina
| | - Jorge F. Elgart
- Centro de Endocrinología Experimental y Aplicada (UNLP-CONICET-CeAs CICPBA), Facultad de Ciencias Médicas UNLP, La Plata, Argentina
| | - Analía G. Abraham
- Centro de Investigación y Desarrollo en Criotecnología de Alimentos, CIDCA (CONICET- UNLP- CIC.PBA), La Plata, Argentina
- Área Bioquímica y Control de Alimentos, Facultad de Ciencias Exactas- UNLP, La Plata, Argentina
| | - Graciela L. Garrote
- Centro de Investigación y Desarrollo en Criotecnología de Alimentos, CIDCA (CONICET- UNLP- CIC.PBA), La Plata, Argentina
| | - Rocío Torrieri
- Centro de Endocrinología Experimental y Aplicada (UNLP-CONICET-CeAs CICPBA), Facultad de Ciencias Médicas UNLP, La Plata, Argentina
| | - Alberto Cepeda
- Laboratorio de Higiene, Inspección y Control de Alimentos (LHICA). Departamento de Química Analítica, Nutrición y Bromatología. Facultad de Veterinaria, Universidade de Santiago de Compostela, Campus de Lugo, Lugo, Spain
| | - Alejandra Cardelle-Cobas
- Laboratorio de Higiene, Inspección y Control de Alimentos (LHICA). Departamento de Química Analítica, Nutrición y Bromatología. Facultad de Veterinaria, Universidade de Santiago de Compostela, Campus de Lugo, Lugo, Spain
| | - Juan J. Gagliardino
- Centro de Endocrinología Experimental y Aplicada (UNLP-CONICET-CeAs CICPBA), Facultad de Ciencias Médicas UNLP, La Plata, Argentina
| |
Collapse
|
44
|
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
|
45
|
Chandrashekar R, Rai M, Kalal BS. Acute and chronic toxicity studies on ethanolic leaf extracts of Clerodendrum viscosum and Leucas indica in Swiss albino mice. INTERNATIONAL JOURNAL OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2022; 13:40-48. [PMID: 36188728 PMCID: PMC9520248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/16/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND AND OBJECTIVES To evaluate the safe dose range of Clerodendrum viscosum (C. viscosum) and Leucas indica (L. indica) ethanolic leaf extracts of acute and chronic oral toxicity study in Swiss Albino mice. MATERIALS AND METHODS The Organization for Economic Co-operation and Development guideline was used for the toxicity studies. C. viscosum and L. indica plant extract were administered orally in a single dose of 2000 mg/kg, and general behavior, adverse effects, and mortality were studied for 72 h. For the chronic toxicity study, both plant extracts were administered orally to a separate set of animals at 300 mg/kg doses for 90 days. Animals body weight was taken out, blood and gastric juice were collected for biochemical parameters, and vital organs were collected for histopathological studies after sacrificing test and control group animals. RESULTS Both in acute and chronic toxicity assay, there was no significant alteration in body weight, physical signs, symptoms, hematological, biochemical parameters, and body organ weights compared to the normal group. The liver, kidney, and stomach histology did not show any drug-induced lesion. CONCLUSIONS The result indicates that the oral administration of C. viscosum and L. indica ethanolic plant extract did not cause any toxicological effects. Hence it could be regarded as a safe natural product for therapeutic use.
Collapse
Affiliation(s)
- Rajan Chandrashekar
- Department of Pharmacology, A. J. Institute of Medical Sciences and Research CentreMangaluru 575004, Karnataka, India
| | - Mohandas Rai
- Department of Pharmacology, A. J. Institute of Medical Sciences and Research CentreMangaluru 575004, Karnataka, India
| | - Bhuvanesh Sukhlal Kalal
- Department of Pharmacology, A. J. Institute of Medical Sciences and Research CentreMangaluru 575004, Karnataka, India
- A. J. Research Centre, A. J. Institute of Medical Sciences & Research CentreMangaluru 575004, Karnataka, India
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of KentuckyLexington, Kentucky 40536, USA
| |
Collapse
|
46
|
Morris EJ, Hollmann J, Hofer AK, Bhagwandass H, Oueini R, Adkins LE, Hallas J, Vouri SM. Evaluating the use of prescription sequence symmetry analysis as a pharmacovigilance tool: A scoping review. Res Social Adm Pharm 2022; 18:3079-3093. [PMID: 34376366 DOI: 10.1016/j.sapharm.2021.08.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/25/2021] [Accepted: 08/03/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND The (prescription) sequence symmetry analysis (PSSA) design has been used to identify potential prescribing cascade signals by assessing the prescribing sequence of an index drug relative to a marker drug presumed to treat an adverse drug event provoked by the index drug. OBJECTIVES This review aimed to explore the use of the PSSA design as a pharmacovigilance tool with a particular focus on the breadth of identified signals and advances in PSSA methodology. METHODS We searched Embase, PubMed/Medline, Google Scholar, Web of Science and grey literature to identify studies that used the PSSA methodology. Two reviewers independently extracted relevant data for each included article. Study characteristics including signals identified, exposure time window, stratified analyses, and use of controls were extracted. RESULTS We identified 53 studies which reported original results obtained using PSSA methodology or quantified the validity of components of the PSSA design. Of those, nine studies provided validation metrics showing reasonable sensitivity and high specificity of PSSA to identify prescribing cascade signals. We identified 340 unique index drug - marker drug signals published in the PSSA literature, representing 281 unique index - marker pharmacological class dyads (i.e., unique fourth-level Anatomical Therapeutic Chemical [ATC] classification dyads). Commonly observed signals were identified for index drugs acting upon the nervous system (34%), cardiovascular system (21%), and blood and blood-forming organs (15%), and many marker drugs were related to the nervous system (25%), alimentary tract and metabolism (23%), cardiovascular system (17%), and genitourinary system and sex hormones (14%). Negative controls and positive controls were utilized in 21% and 13% of studies, respectively. CONCLUSIONS The PSSA methodology has been used in 53 studies worldwide to detect and evaluate over 300 unique prescribing cascades signals. Researchers should consider sensitivity analyses incorporating negative and/or positive controls and additional time windows to evaluate time-varying biases when designing PSSA studies.
Collapse
Affiliation(s)
- Earl J Morris
- Department of Pharmaceutical Outcomes and Policy, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Josef Hollmann
- Department of Pharmaceutical Outcomes and Policy, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Ann-Kathrin Hofer
- Department of Pharmaceutical Outcomes and Policy, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Hemita Bhagwandass
- Department of Pharmaceutical Outcomes and Policy, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Razanne Oueini
- Department of Pharmaceutical Outcomes and Policy, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Lauren E Adkins
- Health Science Center Libraries, University of Florida, Gainesville, FL, USA
| | - Jesper Hallas
- Clinical Pharmacology and Pharmacy, IST, University of Southern Denmark, Odense, Denmark; Department of Clinical Pharmacology and Biochemistry, Odense University Hospital, Odense, Denmark
| | - Scott M Vouri
- Department of Pharmaceutical Outcomes and Policy, College of Pharmacy, University of Florida, Gainesville, FL, USA; Center for Drug Evaluation and Safety, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
47
|
Discovery of Therapeutics Targeting Oxidative Stress in Autosomal Recessive Cerebellar Ataxia: A Systematic Review. Pharmaceuticals (Basel) 2022; 15:ph15060764. [PMID: 35745683 PMCID: PMC9228961 DOI: 10.3390/ph15060764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/05/2022] [Accepted: 06/14/2022] [Indexed: 01/05/2023] Open
Abstract
Autosomal recessive cerebellar ataxias (ARCAs) are a heterogeneous group of rare neurodegenerative inherited disorders. The resulting motor incoordination and progressive functional disabilities lead to reduced lifespan. There is currently no cure for ARCAs, likely attributed to the lack of understanding of the multifaceted roles of antioxidant defense and the underlying mechanisms. This systematic review aims to evaluate the extant literature on the current developments of therapeutic strategies that target oxidative stress for the management of ARCAs. We searched PubMed, Web of Science, and Science Direct Scopus for relevant peer-reviewed articles published from 1 January 2016 onwards. A total of 28 preclinical studies fulfilled the eligibility criteria for inclusion in this systematic review. We first evaluated the altered cellular processes, abnormal signaling cascades, and disrupted protein quality control underlying the pathogenesis of ARCA. We then examined the current potential therapeutic strategies for ARCAs, including aromatic, organic and pharmacological compounds, gene therapy, natural products, and nanotechnology, as well as their associated antioxidant pathways and modes of action. We then discussed their potential as antioxidant therapeutics for ARCAs, with the long-term view toward their possible translation to clinical practice. In conclusion, our current understanding is that these antioxidant therapies show promise in improving or halting the progression of ARCAs. Tailoring the therapies to specific disease stages could greatly facilitate the management of ARCAs.
Collapse
|
48
|
Chong YJ, Azzopardi M, Tallouzi MO, Spooner D, Masood I, Ghosh Y, Sreekantam S. Bilateral Panuveitis and Exudative Retinal Detachments Associated with Alpelisib. Case Rep Oncol 2022; 15:713-719. [PMID: 36157688 PMCID: PMC9459638 DOI: 10.1159/000525772] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 06/21/2022] [Indexed: 11/29/2022] Open
Abstract
We report a case of alpelisib-induced uveitis. A 68-year-old female who had recently been given alpelisib for metastatic breast cancer presented with a 2-week history of bilateral worsening vision with a corresponding acute hypermetropic shift. Her unaided visual acuity was 6/60 in both eyes, with bilateral anterior uveitis, non-granulomatous keratic precipitates, posterior synechiae, and limited fundal view. There was also a mild iris bombe configuration, although the intraocular pressures were normal. Ocular ultrasound revealed bilateral uveal effusion, ciliary body congestion, dense vitreous cells, and exudative retinal detachments. These findings were also confirmed on multimodal imaging with widefield fundus photography (Optos) and optical coherence tomography. Based on the clinical features above, a diagnosis of alpelisib-induced panuveitis was diagnosed. She was then admitted and treated with a 3-day course of intravenous methylprednisolone and intensive topical steroids. Her clinical signs and symptoms started to improve, and she was discharged 4 days later. At 1 week of follow-up, her best-corrected visual acuity was 6/12 in both eyes, with broken posterior synechiae and resolution of exudative retinal detachments. This case highlights the importance of early ophthalmology involvement by the oncology team as oncology therapy can have potential unexpected ocular manifestations.
Collapse
Affiliation(s)
- Yu Jeat Chong
- Birmingham and Midland Eye Centre, Birmingham, United Kingdom
| | | | | | - David Spooner
- Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Imran Masood
- Birmingham and Midland Eye Centre, Birmingham, United Kingdom
| | - Yajati Ghosh
- Birmingham and Midland Eye Centre, Birmingham, United Kingdom
| | | |
Collapse
|
49
|
Xuan P, Wang M, Liu Y, Wang D, Zhang T, Nakaguchi T. Integrating specific and common topologies of heterogeneous graphs and pairwise attributes for drug-related side effect prediction. Brief Bioinform 2022; 23:6573962. [PMID: 35470853 DOI: 10.1093/bib/bbac126] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/15/2022] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
MOTIVATION Computerized methods for drug-related side effect identification can help reduce costs and speed up drug development. Multisource data about drug and side effects are widely used to predict potential drug-related side effects. Heterogeneous graphs are commonly used to associate multisourced data of drugs and side effects which can reflect similarities of the drugs from different perspectives. Effective integration and formulation of diverse similarities, however, are challenging. In addition, the specific topology of each heterogeneous graph and the common topology of multiple graphs are neglected. RESULTS We propose a drug-side effect association prediction model, GCRS, to encode and integrate specific topologies, common topologies and pairwise attributes of drugs and side effects. First, multiple drug-side effect heterogeneous graphs are constructed using various kinds of similarities and associations related to drugs and side effects. As each heterogeneous graph has its specific topology, we establish separate module based on graph convolutional autoencoder (GCA) to learn the particular topology representation of each drug node and each side effect node, respectively. Since multiple graphs reflect the complex relationships among the drug and side effect nodes and contain common topologies, we construct a module based on GCA with sharing parameters to learn the common topology representations of each node. Afterwards, we design an attention mechanism to obtain more informative topology representations at the representation level. Finally, multi-layer convolutional neural networks with attribute-level attention are constructed to deeply integrate the similarity and association attributes of a pair of drug-side effect nodes. Comprehensive experiments show that GCRS's prediction performance is superior to other comparing state-of-the-art methods for predicting drug-side effect associations. The recall rates in top-ranked candidates and case studies on five drugs further demonstrate GCRS's ability in discovering potential drug-related side effects. CONTACT zhang@hlju.edu.cn.
Collapse
Affiliation(s)
- Ping Xuan
- School of Computer Science and Technology, Heilongjiang University, Harbin 150080, China.,School of Computer Science, Shaanxi Normal University, Xi'an 710062, China
| | - Meng Wang
- School of Computer Science and Technology, Heilongjiang University, Harbin 150080, China
| | - Yong Liu
- School of Computer Science and Technology, Heilongjiang University, Harbin 150080, China
| | - Dong Wang
- School of Computer Science and Technology, Heilongjiang University, Harbin 150080, China
| | - Tiangang Zhang
- School of Mathematical Science, Heilongjiang University, Harbin 150080, China
| | - Toshiya Nakaguchi
- Center for Frontier Medical Engineering, Chiba University, Chiba 2638522, Japan
| |
Collapse
|
50
|
Paci P, Fiscon G, Conte F, Wang RS, Handy DE, Farina L, Loscalzo J. Comprehensive network medicine-based drug repositioning via integration of therapeutic efficacy and side effects. NPJ Syst Biol Appl 2022; 8:12. [PMID: 35443763 PMCID: PMC9021283 DOI: 10.1038/s41540-022-00221-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/19/2022] [Indexed: 12/28/2022] Open
Abstract
Despite advances in modern medicine that led to improvements in cardiovascular outcomes, cardiovascular disease (CVD) remains the leading cause of mortality and morbidity globally. Thus, there is an urgent need for new approaches to improve CVD drug treatments. As the development time and cost of drug discovery to clinical application are excessive, alternate strategies for drug development are warranted. Among these are included computational approaches based on omics data for drug repositioning, which have attracted increasing attention. In this work, we developed an adjusted similarity measure implemented by the algorithm SAveRUNNER to reposition drugs for cardiovascular diseases while, at the same time, considering the side effects of drug candidates. We analyzed nine cardiovascular disorders and two side effects. We formulated both disease disorders and side effects as network modules in the human interactome, and considered those drug candidates that are proximal to disease modules but far from side-effects modules as ideal. Our method provides a list of drug candidates for cardiovascular diseases that are unlikely to produce common, adverse side-effects. This approach incorporating side effects is applicable to other diseases, as well.
Collapse
Affiliation(s)
- Paola Paci
- Department of Computer, Control and Management Engineering, Sapienza University of Rome, Rome, Italy. .,Institute for Systems Analysis and Computer Science "Antonio Ruberti", National Research Council, Rome, Italy.
| | - Giulia Fiscon
- Department of Computer, Control and Management Engineering, Sapienza University of Rome, Rome, Italy.,Institute for Systems Analysis and Computer Science "Antonio Ruberti", National Research Council, Rome, Italy
| | - Federica Conte
- Institute for Systems Analysis and Computer Science "Antonio Ruberti", National Research Council, Rome, Italy
| | - Rui-Sheng Wang
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Diane E Handy
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Lorenzo Farina
- Department of Computer, Control and Management Engineering, Sapienza University of Rome, Rome, Italy
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|