1
|
Shen J, Wang J. Art design integrating visual relation and affective semantics based on Convolutional Block Attention Mechanism-generative adversarial network model. PeerJ Comput Sci 2024; 10:e2274. [PMID: 39314726 PMCID: PMC11419622 DOI: 10.7717/peerj-cs.2274] [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: 06/04/2024] [Accepted: 07/29/2024] [Indexed: 09/25/2024]
Abstract
Scene-based image semantic extraction and its precise sentiment expression significantly enhance artistic design. To address the incongruity between image features and sentiment features caused by non-bilinear pooling, this study introduces a generative adversarial network (GAN) model that integrates visual relationships with sentiment semantics. The GAN-based regularizer is utilized during training to incorporate target information derived from the contextual information into the process. This regularization mechanism imposes stronger penalties for inaccuracies in subject-object type predictions and integrates a sentiment corpus to generate more human-like descriptive statements. The capsule network is employed to reconstruct sentences and predict probabilities in the discriminator. To preserve crucial focal points in feature extraction, the Convolutional Block Attention Mechanism (CBAM) is introduced. Furthermore, two bidirectional long short-term memory (LSTM) modules are used to model both target and relational contexts, thereby refining target labels and inter-target relationships. Experimental results highlight the model's superiority over comparative models in terms of accuracy, BiLingual Evaluation Understudy (BLEU) score, and text preservation rate. The proposed model achieves an accuracy of 95.40% and the highest BLEU score of 16.79, effectively capturing both the label content and the emotional nuances within the image.
Collapse
Affiliation(s)
- Jiadong Shen
- School of Design and Art, Changsha University of Science and Technology, Changsha, Hunan, China
| | - Jian Wang
- School of Design and Art, Changsha University of Science and Technology, Changsha, Hunan, China
| |
Collapse
|
2
|
Ishfaq M, Shah SZA, Ahmad I, Rahman Z. Multinomial classification of NLRP3 inhibitory compounds based on large scale machine learning approaches. Mol Divers 2024; 28:1849-1868. [PMID: 37418166 DOI: 10.1007/s11030-023-10690-y] [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: 05/19/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
The role of NLRP3 inflammasome in innate immunity is newly recognized. The NLRP3 protein is a family of nucleotide-binding and oligomerization domain-like receptors as well as a pyrin domain-containing protein. It has been shown that NLRP3 may contribute to the development and progression of a variety of diseases, such as multiple sclerosis, metabolic disorders, inflammatory bowel disease, and other auto-immune and auto-inflammatory conditions. The use of machine learning methods in pharmaceutical research has been widespread for several decades. An important objective of this study is to apply machine learning approaches for the multinomial classification of NLRP3 inhibitors. However, data imbalances can affect machine learning. Therefore, a synthetic minority oversampling technique (SMOTE) has been developed to increase the sensitivity of classifiers to minority groups. The QSAR modelling was performed using 154 molecules retrieved from the ChEMBL database (version 29). The accuracy of the multiclass classification top six models was found to fall within ranges of 0.99 to 0.86, and log loss ranges of 0.2 to 2.3, respectively. The results showed that the receiver operating characteristic curve (ROC) plot values significantly improved when tuning parameters were adjusted and imbalanced data was handled. Moreover, the results demonstrated that SMOTE offers a significant advantage in handling imbalanced datasets as well as substantial improvements in overall accuracy of machine learning models. The top models were then used to predict data from unseen datasets. In summary, these QSAR classification models exhibited robust statistical results and were interpretable, which strongly supported their use for rapid screening of NLRP3 inhibitors.
Collapse
Affiliation(s)
- Muhammad Ishfaq
- College of Computer Science, Huanggang Normal University, Huanggang, 438000, China
| | - Syed Zahid Ali Shah
- Department of Pathology, Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Ijaz Ahmad
- The University of Agriculture Peshawar, Peshawar, 25130, Khyber Pakhtunkhwa, Pakistan
| | - Ziaur Rahman
- College of Computer Science, Huanggang Normal University, Huanggang, 438000, China.
| |
Collapse
|
3
|
Nguyen ATN, Nguyen DTN, Koh HY, Toskov J, MacLean W, Xu A, Zhang D, Webb GI, May LT, Halls ML. The application of artificial intelligence to accelerate G protein-coupled receptor drug discovery. Br J Pharmacol 2024; 181:2371-2384. [PMID: 37161878 DOI: 10.1111/bph.16140] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 04/14/2023] [Accepted: 04/27/2023] [Indexed: 05/11/2023] Open
Abstract
The application of artificial intelligence (AI) approaches to drug discovery for G protein-coupled receptors (GPCRs) is a rapidly expanding area. Artificial intelligence can be used at multiple stages during the drug discovery process, from aiding our understanding of the fundamental actions of GPCRs to the discovery of new ligand-GPCR interactions or the prediction of clinical responses. Here, we provide an overview of the concepts behind artificial intelligence, including the subfields of machine learning and deep learning. We summarise the published applications of artificial intelligence to different stages of the GPCR drug discovery process. Finally, we reflect on the benefits and limitations of artificial intelligence and share our vision for the exciting potential for further development of applications to aid GPCR drug discovery. In addition to making the drug discovery process "faster, smarter and cheaper," we anticipate that the application of artificial intelligence will create exciting new opportunities for GPCR drug discovery. LINKED ARTICLES: This article is part of a themed issue Therapeutic Targeting of G Protein-Coupled Receptors: hot topics from the Australasian Society of Clinical and Experimental Pharmacologists and Toxicologists 2021 Virtual Annual Scientific Meeting. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.14/issuetoc.
Collapse
Affiliation(s)
- Anh T N Nguyen
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Diep T N Nguyen
- Department of Information Technology, Faculty of Engineering and Technology, Vietnam National University, Cau Giay, Hanoi, Vietnam
| | - Huan Yee Koh
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Monash Data Futures Institute and Department of Data Science and Artificial Intelligence, Monash University, Clayton, Victoria, Australia
| | - Jason Toskov
- Monash DeepNeuron, Monash University, Clayton, Victoria, Australia
| | - William MacLean
- Monash DeepNeuron, Monash University, Clayton, Victoria, Australia
| | - Andrew Xu
- Monash DeepNeuron, Monash University, Clayton, Victoria, Australia
| | - Daokun Zhang
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Monash Data Futures Institute and Department of Data Science and Artificial Intelligence, Monash University, Clayton, Victoria, Australia
| | - Geoffrey I Webb
- Monash Data Futures Institute and Department of Data Science and Artificial Intelligence, Monash University, Clayton, Victoria, Australia
| | - Lauren T May
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Michelle L Halls
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
4
|
Van Booven DJ, Chen CB, Malpani S, Mirzabeigi Y, Mohammadi M, Wang Y, Kryvenko ON, Punnen S, Arora H. Synthetic Genitourinary Image Synthesis via Generative Adversarial Networks: Enhancing Artificial Intelligence Diagnostic Precision. J Pers Med 2024; 14:703. [PMID: 39063957 PMCID: PMC11278131 DOI: 10.3390/jpm14070703] [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: 05/23/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
INTRODUCTION In the realm of computational pathology, the scarcity and restricted diversity of genitourinary (GU) tissue datasets pose significant challenges for training robust diagnostic models. This study explores the potential of Generative Adversarial Networks (GANs) to mitigate these limitations by generating high-quality synthetic images of rare or underrepresented GU tissues. We hypothesized that augmenting the training data of computational pathology models with these GAN-generated images, validated through pathologist evaluation and quantitative similarity measures, would significantly enhance model performance in tasks such as tissue classification, segmentation, and disease detection. METHODS To test this hypothesis, we employed a GAN model to produce synthetic images of eight different GU tissues. The quality of these images was rigorously assessed using a Relative Inception Score (RIS) of 1.27 ± 0.15 and a Fréchet Inception Distance (FID) that stabilized at 120, metrics that reflect the visual and statistical fidelity of the generated images to real histopathological images. Additionally, the synthetic images received an 80% approval rating from board-certified pathologists, further validating their realism and diagnostic utility. We used an alternative Spatial Heterogeneous Recurrence Quantification Analysis (SHRQA) to assess the quality of prostate tissue. This allowed us to make a comparison between original and synthetic data in the context of features, which were further validated by the pathologist's evaluation. Future work will focus on implementing a deep learning model to evaluate the performance of the augmented datasets in tasks such as tissue classification, segmentation, and disease detection. This will provide a more comprehensive understanding of the utility of GAN-generated synthetic images in enhancing computational pathology workflows. RESULTS This study not only confirms the feasibility of using GANs for data augmentation in medical image analysis but also highlights the critical role of synthetic data in addressing the challenges of dataset scarcity and imbalance. CONCLUSIONS Future work will focus on refining the generative models to produce even more diverse and complex tissue representations, potentially transforming the landscape of medical diagnostics with AI-driven solutions.
Collapse
Affiliation(s)
- Derek J. Van Booven
- John P Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Cheng-Bang Chen
- Department of Industrial and Systems Engineering, University of Miami, Coral Gables, FL 33146, USA; (C.-B.C.); (Y.W.)
| | - Sheetal Malpani
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (S.M.); (Y.M.); (O.N.K.)
| | - Yasamin Mirzabeigi
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (S.M.); (Y.M.); (O.N.K.)
| | - Maral Mohammadi
- Department of Pathology, University of Debrecen in Hungary, 4032 Debrecen, Hungary;
| | - Yujie Wang
- Department of Industrial and Systems Engineering, University of Miami, Coral Gables, FL 33146, USA; (C.-B.C.); (Y.W.)
| | - Oleksander N. Kryvenko
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (S.M.); (Y.M.); (O.N.K.)
| | - Sanoj Punnen
- Desai & Sethi Institute of Urology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Himanshu Arora
- John P Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Department of Pathology, University of Debrecen in Hungary, 4032 Debrecen, Hungary;
- Desai & Sethi Institute of Urology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- The Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
5
|
Wei W, Fang J, Yang N, Li Q, Hu L, Zhao L, Han J. AC-ModNet: Molecular Reverse Design Network Based on Attribute Classification. Int J Mol Sci 2024; 25:6940. [PMID: 39000049 PMCID: PMC11241775 DOI: 10.3390/ijms25136940] [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: 05/12/2024] [Revised: 06/13/2024] [Accepted: 06/22/2024] [Indexed: 07/14/2024] Open
Abstract
Deep generative models are becoming a tool of choice for exploring the molecular space. One important application area of deep generative models is the reverse design of drug compounds for given attributes (solubility, ease of synthesis, etc.). Although there are many generative models, these models cannot generate specific intervals of attributes. This paper proposes a AC-ModNet model that effectively combines VAE with AC-GAN to generate molecular structures in specific attribute intervals. The AC-ModNet is trained and evaluated using the open 250K ZINC dataset. In comparison with related models, our method performs best in the FCD and Frag model evaluation indicators. Moreover, we prove the AC-ModNet created molecules have potential application value in drug design by comparing and analyzing them with medical records in the PubChem database. The results of this paper will provide a new method for machine learning drug reverse design.
Collapse
Affiliation(s)
| | | | - Ning Yang
- School of Automation, Northwestern Polytechnical University, Xi’an 710072, China; (W.W.); (J.F.); (Q.L.); (L.H.); (L.Z.); (J.H.)
| | | | | | | | | |
Collapse
|
6
|
Kyro GW, Morgunov A, Brent RI, Batista VS. ChemSpaceAL: An Efficient Active Learning Methodology Applied to Protein-Specific Molecular Generation. J Chem Inf Model 2024; 64:653-665. [PMID: 38287889 DOI: 10.1021/acs.jcim.3c01456] [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: 01/31/2024]
Abstract
The incredible capabilities of generative artificial intelligence models have inevitably led to their application in the domain of drug discovery. Within this domain, the vastness of chemical space motivates the development of more efficient methods for identifying regions with molecules that exhibit desired characteristics. In this work, we present a computationally efficient active learning methodology and demonstrate its applicability to targeted molecular generation. When applied to c-Abl kinase, a protein with FDA-approved small-molecule inhibitors, the model learns to generate molecules similar to the inhibitors without prior knowledge of their existence and even reproduces two of them exactly. We also show that the methodology is effective for a protein without any commercially available small-molecule inhibitors, the HNH domain of the CRISPR-associated protein 9 (Cas9) enzyme. To facilitate implementation and reproducibility, we made all of our software available through the open-source ChemSpaceAL Python package.
Collapse
Affiliation(s)
- Gregory W Kyro
- Department of Chemistry, Yale University, New Haven, Connecticut 06511-8499, United States
| | - Anton Morgunov
- Department of Chemistry, Yale University, New Haven, Connecticut 06511-8499, United States
| | - Rafael I Brent
- Department of Chemistry, Yale University, New Haven, Connecticut 06511-8499, United States
| | - Victor S Batista
- Department of Chemistry, Yale University, New Haven, Connecticut 06511-8499, United States
| |
Collapse
|
7
|
Gangwal A, Ansari A, Ahmad I, Azad AK, Kumarasamy V, Subramaniyan V, Wong LS. Generative artificial intelligence in drug discovery: basic framework, recent advances, challenges, and opportunities. Front Pharmacol 2024; 15:1331062. [PMID: 38384298 PMCID: PMC10879372 DOI: 10.3389/fphar.2024.1331062] [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: 11/07/2023] [Accepted: 01/17/2024] [Indexed: 02/23/2024] Open
Abstract
There are two main ways to discover or design small drug molecules. The first involves fine-tuning existing molecules or commercially successful drugs through quantitative structure-activity relationships and virtual screening. The second approach involves generating new molecules through de novo drug design or inverse quantitative structure-activity relationship. Both methods aim to get a drug molecule with the best pharmacokinetic and pharmacodynamic profiles. However, bringing a new drug to market is an expensive and time-consuming endeavor, with the average cost being estimated at around $2.5 billion. One of the biggest challenges is screening the vast number of potential drug candidates to find one that is both safe and effective. The development of artificial intelligence in recent years has been phenomenal, ushering in a revolution in many fields. The field of pharmaceutical sciences has also significantly benefited from multiple applications of artificial intelligence, especially drug discovery projects. Artificial intelligence models are finding use in molecular property prediction, molecule generation, virtual screening, synthesis planning, repurposing, among others. Lately, generative artificial intelligence has gained popularity across domains for its ability to generate entirely new data, such as images, sentences, audios, videos, novel chemical molecules, etc. Generative artificial intelligence has also delivered promising results in drug discovery and development. This review article delves into the fundamentals and framework of various generative artificial intelligence models in the context of drug discovery via de novo drug design approach. Various basic and advanced models have been discussed, along with their recent applications. The review also explores recent examples and advances in the generative artificial intelligence approach, as well as the challenges and ongoing efforts to fully harness the potential of generative artificial intelligence in generating novel drug molecules in a faster and more affordable manner. Some clinical-level assets generated form generative artificial intelligence have also been discussed in this review to show the ever-increasing application of artificial intelligence in drug discovery through commercial partnerships.
Collapse
Affiliation(s)
- Amit Gangwal
- Department of Natural Product Chemistry, Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule, Maharashtra, India
| | - Azim Ansari
- Computer Aided Drug Design Center Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule, Maharashtra, India
| | - Iqrar Ahmad
- Department of Pharmaceutical Chemistry, Prof. Ravindra Nikam College of Pharmacy, Dhule, India
| | - Abul Kalam Azad
- Faculty of Pharmacy, University College of MAIWP International, Batu Caves, Malaysia
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Selangor, Malaysia
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Nilai, Malaysia
| |
Collapse
|
8
|
Ivanenkov Y, Zagribelnyy B, Malyshev A, Evteev S, Terentiev V, Kamya P, Bezrukov D, Aliper A, Ren F, Zhavoronkov A. The Hitchhiker's Guide to Deep Learning Driven Generative Chemistry. ACS Med Chem Lett 2023; 14:901-915. [PMID: 37465301 PMCID: PMC10351082 DOI: 10.1021/acsmedchemlett.3c00041] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/09/2023] [Indexed: 07/20/2023] Open
Abstract
This microperspective covers the most recent research outcomes of artificial intelligence (AI) generated molecular structures from the point of view of the medicinal chemist. The main focus is on studies that include synthesis and experimental in vitro validation in biochemical assays of the generated molecular structures, where we analyze the reported structures' relevance in modern medicinal chemistry and their novelty. The authors believe that this review would be appreciated by medicinal chemistry and AI-driven drug design (AIDD) communities and can be adopted as a comprehensive approach for qualifying different research outcomes in AIDD.
Collapse
Affiliation(s)
- Yan Ivanenkov
- Insilico
Medicine Hong Kong Ltd., Science Park East Avenue, Hong Kong Science Park, Pak Shek Kok, Hong Kong
| | - Bogdan Zagribelnyy
- Insilico
Medicine AI Limited, Level 6, Unit 08, Block A, IRENA HQ Building, P.O.
Box 145748, Masdar City, Abu Dhabi United Arab Emirates
| | - Alex Malyshev
- Insilico
Medicine Hong Kong Ltd., Science Park East Avenue, Hong Kong Science Park, Pak Shek Kok, Hong Kong
| | - Sergei Evteev
- Insilico
Medicine Hong Kong Ltd., Science Park East Avenue, Hong Kong Science Park, Pak Shek Kok, Hong Kong
| | - Victor Terentiev
- Insilico
Medicine Hong Kong Ltd., Science Park East Avenue, Hong Kong Science Park, Pak Shek Kok, Hong Kong
| | - Petrina Kamya
- Insilico
Medicine Canada Inc., 3710-1250 René-Lévesque Blvd W, Montreal, Quebec, Canada H3B 4W8
| | - Dmitry Bezrukov
- Insilico
Medicine Hong Kong Ltd., Science Park East Avenue, Hong Kong Science Park, Pak Shek Kok, Hong Kong
| | - Alex Aliper
- Insilico
Medicine AI Limited, Level 6, Unit 08, Block A, IRENA HQ Building, P.O.
Box 145748, Masdar City, Abu Dhabi United Arab Emirates
| | - Feng Ren
- Insilico
Medicine Shanghai Ltd., Suite 901, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong New District, Shanghai 201203, China
| | - Alex Zhavoronkov
- Insilico
Medicine Hong Kong Ltd., Science Park East Avenue, Hong Kong Science Park, Pak Shek Kok, Hong Kong
| |
Collapse
|
9
|
Bian Y, Kwon JJ, Liu C, Margiotta E, Shekhar M, Gould AE. Target-driven machine learning-enabled virtual screening (TAME-VS) platform for early-stage hit identification. Front Mol Biosci 2023; 10:1163536. [PMID: 36994428 PMCID: PMC10040869 DOI: 10.3389/fmolb.2023.1163536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 02/28/2023] [Indexed: 03/15/2023] Open
Abstract
High-throughput screening (HTS) methods enable the empirical evaluation of a large scale of compounds and can be augmented by virtual screening (VS) techniques to save time and money by using potential active compounds for experimental testing. Structure-based and ligand-based virtual screening approaches have been extensively studied and applied in drug discovery practice with proven outcomes in advancing candidate molecules. However, the experimental data required for VS are expensive, and hit identification in an effective and efficient manner is particularly challenging during early-stage drug discovery for novel protein targets. Herein, we present our TArget-driven Machine learning-Enabled VS (TAME-VS) platform, which leverages existing chemical databases of bioactive molecules to modularly facilitate hit finding. Our methodology enables bespoke hit identification campaigns through a user-defined protein target. The input target ID is used to perform a homology-based target expansion, followed by compound retrieval from a large compilation of molecules with experimentally validated activity. Compounds are subsequently vectorized and adopted for machine learning (ML) model training. These machine learning models are deployed to perform model-based inferential virtual screening, and compounds are nominated based on predicted activity. Our platform was retrospectively validated across ten diverse protein targets and demonstrated clear predictive power. The implemented methodology provides a flexible and efficient approach that is accessible to a wide range of users. The TAME-VS platform is publicly available at https://github.com/bymgood/Target-driven-ML-enabled-VS to facilitate early-stage hit identification.
Collapse
Affiliation(s)
- Yuemin Bian
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Jason J. Kwon
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Cong Liu
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Enrico Margiotta
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Mrinal Shekhar
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Alexandra E. Gould
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
10
|
Robust and Fair Undersea Target Detection with Automated Underwater Vehicles for Biodiversity Data Collection. REMOTE SENSING 2022. [DOI: 10.3390/rs14153680] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Undersea/subsea data collection via automated underwater vehicles (AUVs) plays an important role for marine biodiversity research, while it is often much more challenging than the data collection above ground via satellites or AUVs. To enable the automated undersea/subsea data collection system, the AUVs are expected to be able to automatically track the objects of interest through what they can “see” from their mounted underwater cameras, where videos or images could be drastically blurred and degraded in underwater lighting conditions. To solve this challenge, in this work, we propose a cascaded framework by combining a DCGAN (deep convolutional generative adversarial network) with an object detector, i.e., single-shot detector (SSD), named DCGAN+SSD, for the detection of various underwater targets from the mounted camera of an automated underwater vehicle. In our framework, our assumption is that DCGAN can be leveraged to alleviate the impact of underwater conditions and provide the object detector with a better performance for automated AUVs. To optimize the hyperparameters of our models, we applied a particle swarm optimization (PSO)-based strategy to improve the performance of our proposed model. In our experiments, we successfully verified our assumption that the DCGAN+SSD architecture can help improve the object detection toward the undersea conditions and achieve apparently better detection rates over the original SSD detector. Further experiments showed that the PSO-based optimization of our models could further improve the model in object detection toward a more robust and fair performance, making our work a promising solution for tackling the challenges in AUVs.
Collapse
|
11
|
Talluri S. Algorithms for protein design. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 130:1-38. [PMID: 35534105 DOI: 10.1016/bs.apcsb.2022.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Computational Protein Design has the potential to contribute to major advances in enzyme technology, vaccine design, receptor-ligand engineering, biomaterials, nanosensors, and synthetic biology. Although Protein Design is a challenging problem, proteins can be designed by experts in Protein Design, as well as by non-experts whose primary interests are in the applications of Protein Design. The increased accessibility of Protein Design technology is attributable to the accumulated knowledge and experience with Protein Design as well as to the availability of software and online resources. The objective of this review is to serve as a guide to the relevant literature with a focus on the novel methods and algorithms that have been developed or applied for Protein Design, and to assist in the selection of algorithms for Protein Design. Novel algorithms and models that have been introduced to utilize the enormous amount of experimental data and novel computational hardware have the potential for producing substantial increases in the accuracy, reliability and range of applications of designed proteins.
Collapse
Affiliation(s)
- Sekhar Talluri
- Department of Biotechnology, GITAM, Visakhapatnam, India.
| |
Collapse
|
12
|
Serov N, Vinogradov V. Artificial intelligence to bring nanomedicine to life. Adv Drug Deliv Rev 2022; 184:114194. [PMID: 35283223 DOI: 10.1016/j.addr.2022.114194] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 12/13/2022]
Abstract
The technology of drug delivery systems (DDSs) has demonstrated an outstanding performance and effectiveness in production of pharmaceuticals, as it is proved by many FDA-approved nanomedicines that have an enhanced selectivity, manageable drug release kinetics and synergistic therapeutic actions. Nonetheless, to date, the rational design and high-throughput development of nanomaterial-based DDSs for specific purposes is far from a routine practice and is still in its infancy, mainly due to the limitations in scientists' capabilities to effectively acquire, analyze, manage, and comprehend complex and ever-growing sets of experimental data, which is vital to develop DDSs with a set of desired functionalities. At the same time, this task is feasible for the data-driven approaches, high throughput experimentation techniques, process automatization, artificial intelligence (AI) technology, and machine learning (ML) approaches, which is referred to as The Fourth Paradigm of scientific research. Therefore, an integration of these approaches with nanomedicine and nanotechnology can potentially accelerate the rational design and high-throughput development of highly efficient nanoformulated drugs and smart materials with pre-defined functionalities. In this Review, we survey the important results and milestones achieved to date in the application of data science, high throughput, as well as automatization approaches, combined with AI and ML to design and optimize DDSs and related nanomaterials. This manuscript mission is not only to reflect the state-of-art in data-driven nanomedicine, but also show how recent findings in the related fields can transform the nanomedicine's image. We discuss how all these results can be used to boost nanomedicine translation to the clinic, as well as highlight the future directions for the development, data-driven, high throughput experimentation-, and AI-assisted design, as well as the production of nanoformulated drugs and smart materials with pre-defined properties and behavior. This Review will be of high interest to the chemists involved in materials science, nanotechnology, and DDSs development for biomedical applications, although the general nature of the presented approaches enables knowledge translation to many other fields of science.
Collapse
Affiliation(s)
- Nikita Serov
- International Institute "Solution Chemistry of Advanced Materials and Technologies", ITMO University, Saint-Petersburg 191002, Russian Federation
| | - Vladimir Vinogradov
- International Institute "Solution Chemistry of Advanced Materials and Technologies", ITMO University, Saint-Petersburg 191002, Russian Federation.
| |
Collapse
|
13
|
Bian Y, Xie XQ. Artificial Intelligent Deep Learning Molecular Generative Modeling of Scaffold-Focused and Cannabinoid CB2 Target-Specific Small-Molecule Sublibraries. Cells 2022; 11:cells11050915. [PMID: 35269537 PMCID: PMC8909864 DOI: 10.3390/cells11050915] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/26/2022] [Accepted: 02/26/2022] [Indexed: 02/01/2023] Open
Abstract
Design and generation of high-quality target- and scaffold-specific small molecules is an important strategy for the discovery of unique and potent bioactive drug molecules. To achieve this goal, authors have developed the deep-learning molecule generation model (DeepMGM) and applied it for the de novo molecular generation of scaffold-focused small-molecule libraries. In this study, a recurrent neural network (RNN) using long short-term memory (LSTM) units was trained with drug-like molecules to result in a general model (g-DeepMGM). Sampling practices on indole and purine scaffolds illustrate the feasibility of creating scaffold-focused chemical libraries based on machine intelligence. Subsequently, a target-specific model (t-DeepMGM) for cannabinoid receptor 2 (CB2) was constructed following the transfer learning process of known CB2 ligands. Sampling outcomes can present similar properties to the reported active molecules. Finally, a discriminator was trained and attached to the DeepMGM to result in an in silico molecular design-test circle. Medicinal chemistry synthesis and biological validation was performed to further investigate the generation outcome, showing that XIE9137 was identified as a potential allosteric modulator of CB2. This study demonstrates how recent progress in deep learning intelligence can benefit drug discovery, especially in de novo molecular design and chemical library generation.
Collapse
Affiliation(s)
- Yuemin Bian
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, Pharmacometrics & System Pharmacology PharmacoAnalytics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- NIH National Center of Excellence for Computational Drug Abuse Research (CDAR), University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, Pharmacometrics & System Pharmacology PharmacoAnalytics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- NIH National Center of Excellence for Computational Drug Abuse Research (CDAR), University of Pittsburgh, Pittsburgh, PA 15261, USA
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Correspondence:
| |
Collapse
|
14
|
Kang SG, Morrone JA, Weber JK, Cornell WD. Analysis of Training and Seed Bias in Small Molecules Generated with a Conditional Graph-Based Variational Autoencoder─Insights for Practical AI-Driven Molecule Generation. J Chem Inf Model 2022; 62:801-816. [PMID: 35130440 DOI: 10.1021/acs.jcim.1c01545] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The application of deep learning to generative molecule design has shown early promise for accelerating lead series development. However, questions remain concerning how factors like training, data set, and seed bias impact the technology's utility to medicinal and computational chemists. In this work, we analyze the impact of seed and training bias on the output of an activity-conditioned graph-based variational autoencoder (VAE). Leveraging a massive, labeled data set corresponding to the dopamine D2 receptor, our graph-based generative model is shown to excel in producing desired conditioned activities and favorable unconditioned physical properties in generated molecules. We implement an activity-swapping method that allows for the activation, deactivation, or retention of activity of molecular seeds, and we apply independent deep learning classifiers to verify the generative results. Overall, we uncover relationships between noise, molecular seeds, and training set selection across a range of latent-space sampling procedures, providing important insights for practical AI-driven molecule generation.
Collapse
Affiliation(s)
- Seung-Gu Kang
- Computational Biology Center, IBM Thomas J. Watson Research Center, 1101 Kitchawan Road, Yorktown Heights, New York 10594, United States
| | - Joseph A Morrone
- Computational Biology Center, IBM Thomas J. Watson Research Center, 1101 Kitchawan Road, Yorktown Heights, New York 10594, United States
| | - Jeffrey K Weber
- Computational Biology Center, IBM Thomas J. Watson Research Center, 1101 Kitchawan Road, Yorktown Heights, New York 10594, United States
| | - Wendy D Cornell
- Computational Biology Center, IBM Thomas J. Watson Research Center, 1101 Kitchawan Road, Yorktown Heights, New York 10594, United States
| |
Collapse
|
15
|
Qi C, Gao J, Chen K, Shu L, Pearson S. Tea Chrysanthemum Detection by Leveraging Generative Adversarial Networks and Edge Computing. FRONTIERS IN PLANT SCIENCE 2022; 13:850606. [PMID: 35463441 PMCID: PMC9021924 DOI: 10.3389/fpls.2022.850606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/09/2022] [Indexed: 05/16/2023]
Abstract
A high resolution dataset is one of the prerequisites for tea chrysanthemum detection with deep learning algorithms. This is crucial for further developing a selective chrysanthemum harvesting robot. However, generating high resolution datasets of the tea chrysanthemum with complex unstructured environments is a challenge. In this context, we propose a novel tea chrysanthemum - generative adversarial network (TC-GAN) that attempts to deal with this challenge. First, we designed a non-linear mapping network for untangling the features of the underlying code. Then, a customized regularization method was used to provide fine-grained control over the image details. Finally, a gradient diversion design with multi-scale feature extraction capability was adopted to optimize the training process. The proposed TC-GAN was compared with 12 state-of-the-art generative adversarial networks, showing that an optimal average precision (AP) of 90.09% was achieved with the generated images (512 × 512) on the developed TC-YOLO object detection model under the NVIDIA Tesla P100 GPU environment. Moreover, the detection model was deployed into the embedded NVIDIA Jetson TX2 platform with 0.1 s inference time, and this edge computing device could be further developed into a perception system for selective chrysanthemum picking robots in the future.
Collapse
Affiliation(s)
- Chao Qi
- College of Engineering, Nanjing Agricultural University, Nanjing, China
| | - Junfeng Gao
- Lincoln Agri-Robotics Centre, Lincoln Institute for Agri-Food Technology, University of Lincoln, Lincoln, United Kingdom
| | - Kunjie Chen
- College of Engineering, Nanjing Agricultural University, Nanjing, China
- *Correspondence: Kunjie Chen,
| | - Lei Shu
- College of Engineering, Nanjing Agricultural University, Nanjing, China
- Lei Shu,
| | - Simon Pearson
- Lincoln Agri-Robotics Centre, Lincoln Institute for Agri-Food Technology, University of Lincoln, Lincoln, United Kingdom
| |
Collapse
|
16
|
Machine learning & deep learning in data-driven decision making of drug discovery and challenges in high-quality data acquisition in the pharmaceutical industry. Future Med Chem 2021; 14:245-270. [PMID: 34939433 DOI: 10.4155/fmc-2021-0243] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Predicting novel small molecule bioactivities for the target deconvolution, hit-to-lead optimization in drug discovery research, requires molecular representation. Previous reports have demonstrated that machine learning (ML) and deep learning (DL) have substantial implications in virtual screening, peptide synthesis, drug ADMET screening and biomarker discovery. These strategies can increase the positive outcomes in the drug discovery process without false-positive rates and can be achieved in a cost-effective way with a minimum duration of time by high-quality data acquisition. This review substantially discusses the recent updates in AI tools as cheminformatics application in medicinal chemistry for the data-driven decision making of drug discovery and challenges in high-quality data acquisition in the pharmaceutical industry while improving small-molecule bioactivities and properties.
Collapse
|
17
|
Joshi RP, Kumar N. Artificial Intelligence for Autonomous Molecular Design: A Perspective. Molecules 2021; 26:6761. [PMID: 34833853 PMCID: PMC8619999 DOI: 10.3390/molecules26226761] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/23/2021] [Accepted: 10/29/2021] [Indexed: 11/23/2022] Open
Abstract
Domain-aware artificial intelligence has been increasingly adopted in recent years to expedite molecular design in various applications, including drug design and discovery. Recent advances in areas such as physics-informed machine learning and reasoning, software engineering, high-end hardware development, and computing infrastructures are providing opportunities to build scalable and explainable AI molecular discovery systems. This could improve a design hypothesis through feedback analysis, data integration that can provide a basis for the introduction of end-to-end automation for compound discovery and optimization, and enable more intelligent searches of chemical space. Several state-of-the-art ML architectures are predominantly and independently used for predicting the properties of small molecules, their high throughput synthesis, and screening, iteratively identifying and optimizing lead therapeutic candidates. However, such deep learning and ML approaches also raise considerable conceptual, technical, scalability, and end-to-end error quantification challenges, as well as skepticism about the current AI hype to build automated tools. To this end, synergistically and intelligently using these individual components along with robust quantum physics-based molecular representation and data generation tools in a closed-loop holds enormous promise for accelerated therapeutic design to critically analyze the opportunities and challenges for their more widespread application. This article aims to identify the most recent technology and breakthrough achieved by each of the components and discusses how such autonomous AI and ML workflows can be integrated to radically accelerate the protein target or disease model-based probe design that can be iteratively validated experimentally. Taken together, this could significantly reduce the timeline for end-to-end therapeutic discovery and optimization upon the arrival of any novel zoonotic transmission event. Our article serves as a guide for medicinal, computational chemistry and biology, analytical chemistry, and the ML community to practice autonomous molecular design in precision medicine and drug discovery.
Collapse
Affiliation(s)
| | - Neeraj Kumar
- Computational Biology Group, Biological Science Division, Pacific Northwest National Laboratory, 902 Battelle Blvd, Richland, WA 99352, USA;
| |
Collapse
|
18
|
Basso F, Pezoa R, Varas M, Villalobos M. A deep learning approach for real-time crash prediction using vehicle-by-vehicle data. ACCIDENT; ANALYSIS AND PREVENTION 2021; 162:106409. [PMID: 34600313 DOI: 10.1016/j.aap.2021.106409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/28/2021] [Accepted: 09/20/2021] [Indexed: 06/13/2023]
Abstract
In road safety, real-time crash prediction may play a crucial role in preventing such traffic events. However, much of the research in this line generally uses data aggregated every five or ten minutes. This article proposes a new image-inspired data architecture capable of capturing the microscopic scene of vehicular behavior. In order to achieve this, an accident-prediction model is built for a section of the Autopista Central urban highway in Santiago, Chile, based on the concatenation of multiple-input Convolutional Neural Networks, using both the aggregated standard traffic data and the proposed architecture. Different oversampling methodologies are analyzed to balance the training data, finding that the Deep Convolutional Generative Adversarial Networks technique with random undersampling presents better results when generating synthetic instances that permit maximizing the predictive performance. Computational experiments suggest that this model outperforms other traditional prediction methodologies in terms of AUC and sensitivity values over a range of false positives with greater applicability in real life.
Collapse
Affiliation(s)
- Franco Basso
- School of Industrial Engineering, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile; Instituto Sistemas Complejos de Ingeniería, Chile.
| | - Raúl Pezoa
- Escuela de Ingeniería Industrial, Universidad Diego Portales, Chile.
| | - Mauricio Varas
- Centro de Investigación en Sustentabilidad y Gestión Estratégica de Recursos, Facultad de Ingeniería, Universidad del Desarrollo, Santiago, Chile.
| | - Matías Villalobos
- Escuela de Ingeniería Industrial, Universidad Diego Portales, Chile.
| |
Collapse
|
19
|
Jia P, Pei J, Wang G, Pan X, Zhu Y, Wu Y, Ouyang L. The roles of computer-aided drug synthesis in drug development. GREEN SYNTHESIS AND CATALYSIS 2021. [DOI: 10.1016/j.gresc.2021.11.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
20
|
Sun K, Xuan X, Zhao L, Long J. Improving the Classification Rate of Insulation Defects in Power Cables by Automatic Features Extraction and Sample Augmentation of Partial Discharge Signal. INT J PATTERN RECOGN 2021. [DOI: 10.1142/s021800142150035x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Conventional pattern recognition methods employed for differentiating the types of insulation defects in power cables usually rely on the manual extraction of partial discharge features, which is inefficient and easily affected by subjective uncertainty. This work addresses this problem by proposing a new framework based on the automatic features extraction of partial discharge signal. The method first applies a sliding time window to convert partial discharge signals in the time domain into two-dimensional images that serve directly as the input to the convolutional neural networks (CNNs). Then a nonlinear encoder is employed to automatically extract the features of the partial discharge image data as the input of CNNs for classification. In addition, we address the overfitting problem associated with the few-shot by applying a deep convolutional generative adversarial network (DCGAN) to augment the original training dataset. Experimental results demonstrate the validity of the proposed algorithm; it increases the classification accuracy by 4.18% relative to that achieved with manually extracted features; the overall accuracy of the proposed algorithm training with the augmented dataset is 3.175% higher than that with the original experimental dataset.
Collapse
Affiliation(s)
- Kang Sun
- School of Electrical Engineering and Automation, Henan Polytechnic University, Jiaozuo, Henan 454000, P. R. China
| | - Xuyang Xuan
- School of Electrical Engineering and Automation, Henan Polytechnic University, Jiaozuo, Henan 454000, P. R. China
| | - Laijun Zhao
- School of Electrical Engineering and Automation, Henan Polytechnic University, Jiaozuo, Henan 454000, P. R. China
| | - Jie Long
- Jiaozuo Power Supply Company, State Grid Henan Electric Power Company, Jiaozuo, Henan 454000, P. R. China
| |
Collapse
|
21
|
Tong X, Liu X, Tan X, Li X, Jiang J, Xiong Z, Xu T, Jiang H, Qiao N, Zheng M. Generative Models for De Novo Drug Design. J Med Chem 2021; 64:14011-14027. [PMID: 34533311 DOI: 10.1021/acs.jmedchem.1c00927] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Artificial intelligence (AI) is booming. Among various AI approaches, generative models have received much attention in recent years. Inspired by these successes, researchers are now applying generative model techniques to de novo drug design, which has been considered as the "holy grail" of drug discovery. In this Perspective, we first focus on describing models such as recurrent neural network, autoencoder, generative adversarial network, transformer, and hybrid models with reinforcement learning. Next, we summarize the applications of generative models to drug design, including generating various compounds to expand the compound library and designing compounds with specific properties, and we also list a few publicly available molecular design tools based on generative models which can be used directly to generate molecules. In addition, we also introduce current benchmarks and metrics frequently used for generative models. Finally, we discuss the challenges and prospects of using generative models to aid drug design.
Collapse
Affiliation(s)
- Xiaochu Tong
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xiaohong Liu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xiaoqin Tan
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xutong Li
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Jiaxin Jiang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Zhaoping Xiong
- Laboratory of Health Intelligence, Huawei Technologies Co., Ltd, Shenzhen 518100, China
| | | | - Hualiang Jiang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Nan Qiao
- Laboratory of Health Intelligence, Huawei Technologies Co., Ltd, Shenzhen 518100, China
| | - Mingyue Zheng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| |
Collapse
|
22
|
Nakaji K, Yamamoto N. Quantum semi-supervised generative adversarial network for enhanced data classification. Sci Rep 2021; 11:19649. [PMID: 34608219 PMCID: PMC8490428 DOI: 10.1038/s41598-021-98933-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 09/16/2021] [Indexed: 11/09/2022] Open
Abstract
In this paper, we propose the quantum semi-supervised generative adversarial network (qSGAN). The system is composed of a quantum generator and a classical discriminator/classifier (D/C). The goal is to train both the generator and the D/C, so that the latter may get a high classification accuracy for a given dataset. Hence the qSGAN needs neither any data loading nor to generate a pure quantum state, implying that qSGAN is much easier to implement than many existing quantum algorithms. Also the generator can serve as a stronger adversary than a classical one thanks to its rich expressibility, and it is expected to be robust against noise. These advantages are demonstrated in a numerical simulation.
Collapse
Affiliation(s)
- Kouhei Nakaji
- Department of Applied Physics and Physico-Informatics and Quantum Computing Center, Keio University, Hiyoshi 3-14-1, Kohoku, Yokohama, 223-8522, Japan.
| | - Naoki Yamamoto
- Department of Applied Physics and Physico-Informatics and Quantum Computing Center, Keio University, Hiyoshi 3-14-1, Kohoku, Yokohama, 223-8522, Japan
| |
Collapse
|
23
|
Hou T, Bian Y, McGuire T, Xie XQ. Integrated Multi-Class Classification and Prediction of GPCR Allosteric Modulators by Machine Learning Intelligence. Biomolecules 2021; 11:biom11060870. [PMID: 34208096 PMCID: PMC8230833 DOI: 10.3390/biom11060870] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/30/2021] [Accepted: 06/08/2021] [Indexed: 01/01/2023] Open
Abstract
G-protein-coupled receptors (GPCRs) are the largest and most diverse group of cell surface receptors that respond to various extracellular signals. The allosteric modulation of GPCRs has emerged in recent years as a promising approach for developing target-selective therapies. Moreover, the discovery of new GPCR allosteric modulators can greatly benefit the further understanding of GPCR cell signaling mechanisms. It is critical but also challenging to make an accurate distinction of modulators for different GPCR groups in an efficient and effective manner. In this study, we focus on an 11-class classification task with 10 GPCR subtype classes and a random compounds class. We used a dataset containing 34,434 compounds with allosteric modulators collected from classical GPCR families A, B, and C, as well as random drug-like compounds. Six types of machine learning models, including support vector machine, naïve Bayes, decision tree, random forest, logistic regression, and multilayer perceptron, were trained using different combinations of features including molecular descriptors, Atom-pair fingerprints, MACCS fingerprints, and ECFP6 fingerprints. The performances of trained machine learning models with different feature combinations were closely investigated and discussed. To the best of our knowledge, this is the first work on the multi-class classification of GPCR allosteric modulators. We believe that the classification models developed in this study can be used as simple and accurate tools for the discovery and development of GPCR allosteric modulators.
Collapse
Affiliation(s)
- Tianling Hou
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screen (CCGS) Center and Pharmacometrics System Pharmacology Program, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (T.H.); (Y.B.); (T.M.)
- NIH National Center of Excellence for Computational Drug Abuse Research (CDAR), University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Yuemin Bian
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screen (CCGS) Center and Pharmacometrics System Pharmacology Program, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (T.H.); (Y.B.); (T.M.)
- NIH National Center of Excellence for Computational Drug Abuse Research (CDAR), University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Terence McGuire
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screen (CCGS) Center and Pharmacometrics System Pharmacology Program, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (T.H.); (Y.B.); (T.M.)
- NIH National Center of Excellence for Computational Drug Abuse Research (CDAR), University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screen (CCGS) Center and Pharmacometrics System Pharmacology Program, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; (T.H.); (Y.B.); (T.M.)
- Drug Discovery Institute, Departments of Computational Biology and of Structural Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Correspondence:
| |
Collapse
|
24
|
Mercado R, Rastemo T, Lindelöf E, Klambauer G, Engkvist O, Chen H, Jannik Bjerrum E. Graph networks for molecular design. MACHINE LEARNING-SCIENCE AND TECHNOLOGY 2021. [DOI: 10.1088/2632-2153/abcf91] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
25
|
Bian Y, Xie XQ. Generative chemistry: drug discovery with deep learning generative models. J Mol Model 2021; 27:71. [PMID: 33543405 PMCID: PMC10984615 DOI: 10.1007/s00894-021-04674-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/13/2021] [Indexed: 12/15/2022]
Abstract
The de novo design of molecular structures using deep learning generative models introduces an encouraging solution to drug discovery in the face of the continuously increased cost of new drug development. From the generation of original texts, images, and videos, to the scratching of novel molecular structures the creativity of deep learning generative models exhibits the height machine intelligence can achieve. The purpose of this paper is to review the latest advances in generative chemistry which relies on generative modeling to expedite the drug discovery process. This review starts with a brief history of artificial intelligence in drug discovery to outline this emerging paradigm. Commonly used chemical databases, molecular representations, and tools in cheminformatics and machine learning are covered as the infrastructure for generative chemistry. The detailed discussions on utilizing cutting-edge generative architectures, including recurrent neural network, variational autoencoder, adversarial autoencoder, and generative adversarial network for compound generation are focused. Challenges and future perspectives follow.
Collapse
Affiliation(s)
- Yuemin Bian
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- NIH National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
- NIH National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
- Drug Discovery Institute, University of Pittsburgh, 335 Sutherland Drive, 206 Salk Pavilion, Pittsburgh, PA, 15261, USA.
- Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, PA, 15261, Pittsburgh, USA.
| |
Collapse
|
26
|
Lin E, Lin CH, Lane HY. Relevant Applications of Generative Adversarial Networks in Drug Design and Discovery: Molecular De Novo Design, Dimensionality Reduction, and De Novo Peptide and Protein Design. Molecules 2020; 25:E3250. [PMID: 32708785 PMCID: PMC7397124 DOI: 10.3390/molecules25143250] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/11/2020] [Accepted: 07/14/2020] [Indexed: 01/16/2023] Open
Abstract
A growing body of evidence now suggests that artificial intelligence and machine learning techniques can serve as an indispensable foundation for the process of drug design and discovery. In light of latest advancements in computing technologies, deep learning algorithms are being created during the development of clinically useful drugs for treatment of a number of diseases. In this review, we focus on the latest developments for three particular arenas in drug design and discovery research using deep learning approaches, such as generative adversarial network (GAN) frameworks. Firstly, we review drug design and discovery studies that leverage various GAN techniques to assess one main application such as molecular de novo design in drug design and discovery. In addition, we describe various GAN models to fulfill the dimension reduction task of single-cell data in the preclinical stage of the drug development pipeline. Furthermore, we depict several studies in de novo peptide and protein design using GAN frameworks. Moreover, we outline the limitations in regard to the previous drug design and discovery studies using GAN models. Finally, we present a discussion of directions and challenges for future research.
Collapse
Affiliation(s)
- Eugene Lin
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA;
- Department of Electrical & Computer Engineering, University of Washington, Seattle, WA 98195, USA
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Chieh-Hsin Lin
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- School of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Hsien-Yuan Lane
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Department of Psychiatry, China Medical University Hospital, Taichung 40447, Taiwan
- Brain Disease Research Center, China Medical University Hospital, Taichung 40447, Taiwan
- Department of Psychology, College of Medical and Health Sciences, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
27
|
Chen Y, Bian Y, Wang JW, Gong TT, Ying YM, Ma LF, Shan WG, Xie XQ, Zhan ZJ. Effects of α-Mangostin Derivatives on the Alzheimer's Disease Model of Rats and Their Mechanism: A Combination of Experimental Study and Computational Systems Pharmacology Analysis. ACS OMEGA 2020; 5:9846-9863. [PMID: 32391472 PMCID: PMC7203693 DOI: 10.1021/acsomega.0c00057] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 04/16/2020] [Indexed: 06/09/2023]
Abstract
α-Mangostin (α-M) is a natural xanthone from the pericarp of fruit Garcinia mangostana and possesses versatile biological activities. α-M has a therapeutic potential to treat Alzheimer's disease (AD) because of its anti-inflammatory, antioxidative, and neuroprotective activities. However, the use of α-M for AD treatment is limited due to its cytotoxic activities and relatively low potency. Modifications of its chemical structure were needed to reduce its cytotoxicity and improve its therapeutic potential against AD. For this purpose, 16 α-M carbamate derivatives were synthesized. An animal model of AD was established, and the effects of AMG-1 on the spatial learning ability and memory ability were evaluated using behavioral tests. The effect on neuropathology was tested by histopathological evaluation, Nissl staining, and silver staining. Computational systems pharmacology analysis using the chemogenomics knowledgebase was applied for network studies. Compound-target, target-pathway, and target-disease networks were constructed, integrating both in silico analysis and reported experimental data. The results show that AMG-1 can demonstrate its therapeutic effects in a one-molecule, multiple-targets manner to remarkably ameliorate neurological changes and reverse behavioral deficits in AD model rats. The improved cognitive function and alleviated neuronal injury can be observed. The ability of AMG-1 to scavenge β-amyloid in the hippocampus was validated in AD model rats.
Collapse
Affiliation(s)
- Yan Chen
- College
of Pharmacology Sciences Zhejiang University of Technology, Hangzhou 310014, P. R. China
- Department of Pharmaceutical
Sciences and Computational Chemical
Genomics Screening Center, School of Pharmacy; NIH National Center of Excellence
for Computational Drug Abuse Research; Drug Discovery Institute; Departments of Computational
Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Yuemin Bian
- Department of Pharmaceutical
Sciences and Computational Chemical
Genomics Screening Center, School of Pharmacy; NIH National Center of Excellence
for Computational Drug Abuse Research; Drug Discovery Institute; Departments of Computational
Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Jian-Wei Wang
- College
of Pharmacology Sciences Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Ting-Ting Gong
- College
of Pharmacology Sciences Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - You-Min Ying
- College
of Pharmacology Sciences Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Lie-Feng Ma
- College
of Pharmacology Sciences Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Wei-Guang Shan
- College
of Pharmacology Sciences Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Xiang-Qun Xie
- Department of Pharmaceutical
Sciences and Computational Chemical
Genomics Screening Center, School of Pharmacy; NIH National Center of Excellence
for Computational Drug Abuse Research; Drug Discovery Institute; Departments of Computational
Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Zha-Jun Zhan
- College
of Pharmacology Sciences Zhejiang University of Technology, Hangzhou 310014, P. R. China
| |
Collapse
|