Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (85)

Search Parameters:
Keywords = computer-aided drug discovery/design

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 495 KB  
Review
Redefining Breast Cancer Care by Harnessing Computational Drug Repositioning
by Elena-Daniela Jurj, Daiana Colibășanu, Sabina-Oana Vasii, Liana Suciu, Cristina Adriana Dehelean and Lucreția Udrescu
Medicina 2025, 61(9), 1640; https://doi.org/10.3390/medicina61091640 - 10 Sep 2025
Viewed by 655
Abstract
Breast cancer faces significant therapeutic challenges, particularly for triple-negative breast cancer (TNBC), due to limited targeted therapies and drug resistance. Drug repositioning leverages existing safety and pharmacokinetic data to expedite the identification of new indications with cost-effective benefits compared to de novo drug [...] Read more.
Breast cancer faces significant therapeutic challenges, particularly for triple-negative breast cancer (TNBC), due to limited targeted therapies and drug resistance. Drug repositioning leverages existing safety and pharmacokinetic data to expedite the identification of new indications with cost-effective benefits compared to de novo drug discovery. In this critical narrative review, we examine recent advances in computational repositioning strategies for breast cancer, focusing on network-based methods, computer-aided drug design, artificial intelligence and machine learning, transcriptomic signature matching, and multi-omics integration. We highlight key case studies that have progressed to preclinical validation or clinical evaluation. We assess comparative performance metrics, experimental validation outcomes, and real-world success rates. We also present critical methodological challenges, including data heterogeneity, bias in real-world data, and the need for study reproducibility. Our review emphasizes the importance of window-of-opportunity trials and the need for standardized data sharing and reproducible pipelines. These insights highlight the groundbreaking potential of in silico repositioning in addressing unmet needs in breast cancer therapy. Full article
(This article belongs to the Special Issue New Developments in Diagnosis and Management of Breast Cancer)
Show Figures

Figure 1

22 pages, 11258 KB  
Article
High-Risk Neuroblastoma Stage 4 (NBS4): Developing a Medicinal Chemistry Multi-Target Drug Approach
by Amgad Gerges and Una Canning
Molecules 2025, 30(10), 2211; https://doi.org/10.3390/molecules30102211 - 19 May 2025
Viewed by 924
Abstract
Childhood neuroblastoma (NB) is a malignant tumour that is a member of a class of embryonic tumours that have their origins in sympathoadrenal progenitor cells. There are five stages in the clinical NB staging system: 1, 2A, 2B, 3, 4S, and 4. For [...] Read more.
Childhood neuroblastoma (NB) is a malignant tumour that is a member of a class of embryonic tumours that have their origins in sympathoadrenal progenitor cells. There are five stages in the clinical NB staging system: 1, 2A, 2B, 3, 4S, and 4. For those diagnosed with stage 4 neuroblastoma (NBS4), the treatment options are limited with a survival rate of between 40 and 50%. Since 1975, more than 15 targets have been identified in the search for a treatment for high-risk NBS4. This article is concerned with the search for a multi-target drug treatment for high-risk NBS4 and focuses on four possible treatment targets that research has identified as having a role in the development of NBS4 and includes the inhibitors Histone Deacetylase (HDAC), Bromodomain (BRD), Hedgehog (HH), and Tropomyosin Kinase (TRK). Computer-aided drug design and molecular modelling have greatly assisted drug discovery in medicinal chemistry. Computational methods such as molecular docking, homology modelling, molecular dynamics, and quantitative structure–activity relationships (QSAR) are frequently used as part of the process for finding new therapeutic drug targets. Relying on these techniques, the authors describe a medicinal chemistry strategy that successfully identified eight compounds (inhibitors) that were thought to be potential inhibitors for each of the four targets listed above. Results revealed that all four targets BRD, HDAC, HH and TRK receptors binding sites share similar amino acid sequencing that ranges from 80 to 100%, offering the possibility of further testing for multi-target drug use. Two additional targets were also tested as part of this work, Retinoic Acid (RA) and c-Src (Csk), which showed similarity (of the binding pocket) across their receptors of 80–100% but lower than 80% for the other four targets. The work for these two targets is the subject of a paper currently in progress. Full article
Show Figures

Graphical abstract

5 pages, 177 KB  
Editorial
Computer-Aided Drug Design and Drug Discovery
by Dragos Paul Mihai and George Mihai Nitulescu
Pharmaceuticals 2025, 18(3), 436; https://doi.org/10.3390/ph18030436 - 20 Mar 2025
Cited by 3 | Viewed by 1533
Abstract
In the rapidly evolving landscape of pharmaceutical research, the integration of computational methods has become a cornerstone in drug discovery and development efforts [...] Full article
(This article belongs to the Section Medicinal Chemistry)
17 pages, 3667 KB  
Review
Drug Discovery for SARS-CoV-2 Utilizing Computer-Aided Drug Design Approaches
by Jiao Guo, Yang Bai, Yan Guo, Meihua Wang, Xinxin Ji and Yang Wang
COVID 2025, 5(3), 32; https://doi.org/10.3390/covid5030032 - 26 Feb 2025
Cited by 1 | Viewed by 1199
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a positive-sense RNA virus with an unusually large genome of approximately 30 kb. It is highly transmissible and exhibits broad tissue tropism. The third most pathogenic of all known coronaviruses, severe acute respiratory syndrome coronavirus [...] Read more.
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a positive-sense RNA virus with an unusually large genome of approximately 30 kb. It is highly transmissible and exhibits broad tissue tropism. The third most pathogenic of all known coronaviruses, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is responsible for the clinical manifestation known as coronavirus disease 2019 (COVID-19), which has resulted in the loss of millions of lives on a global scale. This pandemic has prompted significant efforts to develop therapeutic strategies that target the virus and/or human proteins to control viral infection. These efforts include the testing of hundreds of potential drugs and thousands of patients in clinical trials. Although the global pandemic caused by the SARS-CoV-2 virus is approaching its end, the emergence of new variants and drug-resistant mutants highlights the need for additional oral antivirals. The appearance of variants and the declining effectiveness of booster shots are resulting in breakthrough infections, which continue to impose a significant burden on healthcare systems. Computer-aided drug design (CADD) has been widely utilized for predicting drug–target interactions and evaluating drug safety; it is regarded as an effective tool for identifying promising drug candidates to combat SARS-CoV-2. The CADD approach aids in the discovery of new drugs or the repurposing of United States Food and Drug Administration (FDA)-approved drugs, whose safety and side effects are already well established, thus making the process more viable. This review summarizes potential therapeutic agents that target SARS-CoV-2 or host proteins critical for viral pathogenesis, as identified using CADD approaches. Additionally, this study provides insights into the common in silico methods used in CADD and their current applications in the SARS-CoV-2 drug discovery process. Full article
(This article belongs to the Section COVID Clinical Manifestations and Management)
Show Figures

Figure 1

39 pages, 2406 KB  
Review
A (Comprehensive) Review of the Application of Quantitative Structure–Activity Relationship (QSAR) in the Prediction of New Compounds with Anti-Breast Cancer Activity
by Boris Vasilev and Mariyana Atanasova
Appl. Sci. 2025, 15(3), 1206; https://doi.org/10.3390/app15031206 - 24 Jan 2025
Cited by 9 | Viewed by 10312
Abstract
Computational approaches applied in drug discovery have advanced significantly over the past few decades. These techniques are commonly grouped under the term “computer-aided drug design” (CADD) and are now considered one of the key pillars of pharmaceutical discovery pipelines in both academic and [...] Read more.
Computational approaches applied in drug discovery have advanced significantly over the past few decades. These techniques are commonly grouped under the term “computer-aided drug design” (CADD) and are now considered one of the key pillars of pharmaceutical discovery pipelines in both academic and industrial settings. In this work, we review Quantitative Structure–Activity Relationships (QSARs), one of the most used ligand-based drug design (LBDD) methods, with a focus on its application in the discovery and development of anti-breast cancer drugs. Critical steps in the QSAR methodology, essential for its correct application—but often overlooked, leading to insignificant or misleading models—are examined. Additionally, current anti-breast cancer treatment strategies were briefly overviewed, along with some targets for future treatments. The review covers QSAR studies from the past five years and includes a discussion of notable works that could serve as models for future applications of this interdisciplinary and complex method and that may help in feature drug design and development. Full article
Show Figures

Figure 1

18 pages, 3376 KB  
Article
Heterogeneous Edge Computing for Molecular Property Prediction with Graph Convolutional Networks
by Mahdieh Grailoo and Jose Nunez-Yanez
Electronics 2025, 14(1), 101; https://doi.org/10.3390/electronics14010101 - 30 Dec 2024
Cited by 2 | Viewed by 1224
Abstract
Graph-based neural networks have proven to be useful in molecular property prediction, a critical component of computer-aided drug discovery. In this application, in response to the growing demand for improved computational efficiency and localized edge processing, this paper introduces a novel approach that [...] Read more.
Graph-based neural networks have proven to be useful in molecular property prediction, a critical component of computer-aided drug discovery. In this application, in response to the growing demand for improved computational efficiency and localized edge processing, this paper introduces a novel approach that leverages specialized accelerators on a heterogeneous edge computing platform. Our focus is on graph convolutional networks, a leading graph-based neural network variant that integrates graph convolution layers with multi-layer perceptrons. Molecular graphs are typically characterized by a low number of nodes, leading to low-dimensional dense matrix multiplications within multi-layer perceptrons—conditions that are particularly well-suited for Edge TPUs. These TPUs feature a systolic array of multiply–accumulate units optimized for dense matrix operations. Furthermore, the inherent sparsity in molecular graph adjacency matrices offers additional opportunities for computational optimization. To capitalize on this, we developed an FPGA GFADES accelerator, using high-level synthesis, specifically tailored to efficiently manage the sparsity in both the graph structure and node features. Our hardware/software co-designed GCN+MLP architecture delivers performance improvements, achieving up to 58× increased speed compared to conventional software implementations. This architecture is implemented using the Pynq framework and TensorFlow Lite Runtime, running on a multi-core ARM CPU within an AMD/Xilinx Zynq Ultrascale+ device, in combination with the Edge TPU and programmable logic. Full article
Show Figures

Figure 1

25 pages, 3191 KB  
Article
Machine Learning-Assisted Drug Repurposing Framework for Discovery of Aurora Kinase B Inhibitors
by George Nicolae Daniel Ion, George Mihai Nitulescu and Dragos Paul Mihai
Pharmaceuticals 2025, 18(1), 13; https://doi.org/10.3390/ph18010013 - 25 Dec 2024
Cited by 2 | Viewed by 1946
Abstract
Background: Aurora kinase B (AurB) is a pivotal regulator of mitosis, making it a compelling target for cancer therapy. Despite significant advances in protein kinase inhibitor development, there are currently no AurB inhibitors readily available for therapeutic use. Methods: This study introduces a [...] Read more.
Background: Aurora kinase B (AurB) is a pivotal regulator of mitosis, making it a compelling target for cancer therapy. Despite significant advances in protein kinase inhibitor development, there are currently no AurB inhibitors readily available for therapeutic use. Methods: This study introduces a machine learning-assisted drug repurposing framework integrating quantitative structure-activity relationship (QSAR) modeling, molecular fingerprints-based classification, molecular docking, and molecular dynamics (MD) simulations. Using this pipeline, we analyzed 4680 investigational and approved drugs from DrugBank database. Results: The machine learning models trained for drug repurposing showed satisfying performance and yielded the identification of saredutant, montelukast, and canertinib as potential AurB inhibitors. The candidates demonstrated strong binding energies, key molecular interactions with critical residues (e.g., Phe88, Glu161), and stable MD trajectories, particularly saredutant, a neurokinin-2 (NK2) antagonist. Conclusions: Beyond identifying potential AurB inhibitors, this study highlights an integrated methodology that can be applied to other challenging drug targets. Full article
(This article belongs to the Special Issue Computer-Aided Drug Design and Drug Discovery)
Show Figures

Graphical abstract

11 pages, 2439 KB  
Article
AISMPred: A Machine Learning Approach for Predicting Anti-Inflammatory Small Molecules
by Subathra Selvam, Priya Dharshini Balaji, Honglae Sohn and Thirumurthy Madhavan
Pharmaceuticals 2024, 17(12), 1693; https://doi.org/10.3390/ph17121693 - 15 Dec 2024
Cited by 3 | Viewed by 1960
Abstract
Background/Objectives: Inflammation serves as a vital response to diverse harmful stimuli like infections, toxins, or tissue injuries, aiding in the elimination of pathogens and tissue repair. However, persistent inflammation can lead to chronic diseases. Peptide therapeutics have gained attention for their specificity in [...] Read more.
Background/Objectives: Inflammation serves as a vital response to diverse harmful stimuli like infections, toxins, or tissue injuries, aiding in the elimination of pathogens and tissue repair. However, persistent inflammation can lead to chronic diseases. Peptide therapeutics have gained attention for their specificity in targeting cells, yet their development remains costly and time-consuming. Therefore, small molecules, with their stability, low immunogenicity, and oral bioavailability, have become a focal point for predicting anti-inflammatory small molecules (AISMs). Methods: In this study, we introduce a computational method called AISMPred, designed to classify AISMs and non-AISMs. To develop this approach, we constructed a dataset comprising 1750 AISMs and non-AISMs, each annotated with IC50 values sourced from the PubChem BioAssay database. We computed two distinct types of molecular descriptors using PaDEL and Mordred tools. Subsequently, these descriptors were concatenated to form a hybrid feature set. The SVC-L1 regularization method was implemented for the optimum feature selection to develop robust Machine learning (ML) models. Five different conventional ML classifiers were employed, such as RF, ET, KNN, LR, and Ensemble methods. Results: A total of 15 ML models were developed using 2D, FP, and Hybrid feature sets, with the ET model with hybrid features achieving the highest accuracy of 92% and an AUC of 0.97 on the independent test dataset. Conclusions: This study provides an effective method for screening AISMs, potentially impacting drug discovery and design. Full article
Show Figures

Figure 1

21 pages, 1245 KB  
Perspective
Artificial Intelligence–Driven Computational Approaches in the Development of Anticancer Drugs
by Pankaj Garg, Gargi Singhal, Prakash Kulkarni, David Horne, Ravi Salgia and Sharad S. Singhal
Cancers 2024, 16(22), 3884; https://doi.org/10.3390/cancers16223884 - 20 Nov 2024
Cited by 14 | Viewed by 4525
Abstract
The integration of AI has revolutionized cancer drug development, transforming the landscape of drug discovery through sophisticated computational techniques. AI-powered models and algorithms have enhanced computer-aided drug design (CADD), offering unprecedented precision in identifying potential anticancer compounds. Traditionally, cancer drug design has been [...] Read more.
The integration of AI has revolutionized cancer drug development, transforming the landscape of drug discovery through sophisticated computational techniques. AI-powered models and algorithms have enhanced computer-aided drug design (CADD), offering unprecedented precision in identifying potential anticancer compounds. Traditionally, cancer drug design has been a complex, resource-intensive process, but AI introduces new opportunities to accelerate discovery, reduce costs, and optimize efficiency. This manuscript delves into the transformative applications of AI-driven methodologies in predicting and developing anticancer drugs, critically evaluating their potential to reshape the future of cancer therapeutics while addressing their challenges and limitations. Full article
Show Figures

Figure 1

19 pages, 1846 KB  
Article
Protein Language Models and Machine Learning Facilitate the Identification of Antimicrobial Peptides
by David Medina-Ortiz, Seba Contreras, Diego Fernández, Nicole Soto-García, Iván Moya, Gabriel Cabas-Mora and Álvaro Olivera-Nappa
Int. J. Mol. Sci. 2024, 25(16), 8851; https://doi.org/10.3390/ijms25168851 - 14 Aug 2024
Cited by 8 | Viewed by 3604
Abstract
Peptides are bioactive molecules whose functional versatility in living organisms has led to successful applications in diverse fields. In recent years, the amount of data describing peptide sequences and function collected in open repositories has substantially increased, allowing the application of more complex [...] Read more.
Peptides are bioactive molecules whose functional versatility in living organisms has led to successful applications in diverse fields. In recent years, the amount of data describing peptide sequences and function collected in open repositories has substantially increased, allowing the application of more complex computational models to study the relations between the peptide composition and function. This work introduces AMP-Detector, a sequence-based classification model for the detection of peptides’ functional biological activity, focusing on accelerating the discovery and de novo design of potential antimicrobial peptides (AMPs). AMP-Detector introduces a novel sequence-based pipeline to train binary classification models, integrating protein language models and machine learning algorithms. This pipeline produced 21 models targeting antimicrobial, antiviral, and antibacterial activity, achieving average precision exceeding 83%. Benchmark analyses revealed that our models outperformed existing methods for AMPs and delivered comparable results for other biological activity types. Utilizing the Peptide Atlas, we applied AMP-Detector to discover over 190,000 potential AMPs and demonstrated that it is an integrative approach with generative learning to aid in de novo design, resulting in over 500 novel AMPs. The combination of our methodology, robust models, and a generative design strategy offers a significant advancement in peptide-based drug discovery and represents a pivotal tool for therapeutic applications. Full article
Show Figures

Figure 1

25 pages, 12360 KB  
Article
Identification and Dynamics Understanding of Novel Inhibitors of Peptidase Domain of Collagenase G from Clostridium histolyticum
by Farah Anjum, Ali Hazazi, Fouzeyyah Ali Alsaeedi, Maha Bakhuraysah, Alaa Shafie, Norah Ali Alshehri, Nahed Hawsawi, Amal Adnan Ashour, Hamsa Jameel Banjer, Afaf Alharthi and Maryam Ishrat Niaz
Computation 2024, 12(8), 153; https://doi.org/10.3390/computation12080153 - 25 Jul 2024
Cited by 1 | Viewed by 2233
Abstract
Clostridium histolyticum is a Gram-positive anaerobic bacterium belonging to the Clostridium genus. It produces collagenase, an enzyme involved in breaking down collagen which is a key component of connective tissues. However, antimicrobial resistance (AMR) poses a great challenge in combating infections caused by [...] Read more.
Clostridium histolyticum is a Gram-positive anaerobic bacterium belonging to the Clostridium genus. It produces collagenase, an enzyme involved in breaking down collagen which is a key component of connective tissues. However, antimicrobial resistance (AMR) poses a great challenge in combating infections caused by this bacteria. The lengthy nature of traditional drug development techniques has resulted in a shift to computer-aided drug design and other modern drug discovery approaches. The above method offers a cost-effective means for gathering comprehensive information about how ligands interact with their target proteins. The objective of this study is to create novel, explicit drugs that specifically inhibit the C. histolyticum collagenase enzyme. Through structure-based virtual screening, a library containing 1830 compounds was screened to identify potential drug candidates against collagenase enzymes. Following that, molecular dynamic (MD) simulation was performed in an aqueous solution to evaluate the behavior of protein and ligand in a dynamic environment while density functional theory (DFT) analysis was executed to predict the molecular properties and structure of lead compounds, and the WaterSwap technique was utilized to obtain insights into the drug–protein interaction with water molecules. Furthermore, principal component analysis (PCA) was performed to reveal conformational changes, salt bridges to express electrostatic interaction and protein stability, and absorption, distribution, metabolism, excretion, and toxicity (ADMET) to assess the pharmacokinetics profile of top compounds and control molecules. Three potent drug candidates were identified MSID000001, MSID000002, MSID000003, and the control with a binding score of −10.7 kcal/mol, −9.8 kcal/mol, −9.5 kcal/mol, and −8 kcal/mol, respectively. Furthermore, Molecular Mechanics Poisson–Boltzmann Surface Area (MMPBSA) analysis of the simulation trajectories revealed energy scores of −79.54 kcal/mol, −73.99 kcal/mol, −62.26 kcal/mol, and −70.66 kcal/mol, correspondingly. The pharmacokinetics properties exhibited were under the acceptable range. The compounds hold the potential to be novel drugs; therefore, further investigation needs to be conducted to find out their anti-collagenase action against C. histolyticum infections and antibiotic resistance. Full article
(This article belongs to the Special Issue 10th Anniversary of Computation—Computational Biology)
Show Figures

Figure 1

13 pages, 2719 KB  
Article
Structure-Based Identification of Novel Histone Deacetylase 4 (HDAC4) Inhibitors
by Rupesh Agarwal, Pawat Pattarawat, Michael R. Duff, Hwa-Chain Robert Wang, Jerome Baudry and Jeremy C. Smith
Pharmaceuticals 2024, 17(7), 867; https://doi.org/10.3390/ph17070867 - 2 Jul 2024
Cited by 2 | Viewed by 2465
Abstract
Histone deacetylases (HDACs) are important cancer drug targets. Existing FDA-approved drugs target the catalytic pocket of HDACs, which is conserved across subfamilies (classes) of HDAC. However, engineering specificity is an important goal. Herein, we use molecular modeling approaches to identify and target potential [...] Read more.
Histone deacetylases (HDACs) are important cancer drug targets. Existing FDA-approved drugs target the catalytic pocket of HDACs, which is conserved across subfamilies (classes) of HDAC. However, engineering specificity is an important goal. Herein, we use molecular modeling approaches to identify and target potential novel pockets specific to Class IIA HDAC-HDAC4 at the interface between HDAC4 and the transcriptional corepressor component protein NCoR. These pockets were screened using an ensemble docking approach combined with consensus scoring to identify compounds with a different binding mechanism than the currently known HDAC modulators. Binding was compared in experimental assays between HDAC4 and HDAC3, which belong to a different family of HDACs. HDAC4 was significantly inhibited by compound 88402 but not HDAC3. Two other compounds (67436 and 134199) had IC50 values in the low micromolar range for both HDACs, which is comparable to the known inhibitor of HDAC4, SAHA (Vorinostat). However, both of these compounds were significantly weaker inhibitors of HDAC3 than SAHA and thus more selective, albeit to a limited extent. Five compounds exhibited activity on human breast carcinoma and/or urothelial carcinoma cell lines. The present result suggests potential mechanistic and chemical approaches for developing selective HDAC4 modulators. Full article
(This article belongs to the Special Issue Small Molecule Drug Discovery: Driven by In-Silico Techniques)
Show Figures

Figure 1

19 pages, 7216 KB  
Article
In Silico Design of Potential Small-Molecule Antibiotic Adjuvants against Salmonella typhimurium Ortho Acetyl Sulphydrylase Synthase to Address Antimicrobial Resistance
by Oluwadunni F. Elebiju, Gbolahan O. Oduselu, Temitope A. Ogunnupebi, Olayinka O. Ajani and Ezekiel Adebiyi
Pharmaceuticals 2024, 17(5), 543; https://doi.org/10.3390/ph17050543 - 23 Apr 2024
Cited by 5 | Viewed by 2739
Abstract
The inhibition of O-acetyl sulphydrylase synthase isoforms has been reported to represent a promising approach for the development of antibiotic adjuvants. This occurs via the organism developing an unpaired oxidative stress response, causing a reduction in antibiotic resistance in vegetative and swarm [...] Read more.
The inhibition of O-acetyl sulphydrylase synthase isoforms has been reported to represent a promising approach for the development of antibiotic adjuvants. This occurs via the organism developing an unpaired oxidative stress response, causing a reduction in antibiotic resistance in vegetative and swarm cell populations. This consequently increases the effectiveness of conventional antibiotics at lower doses. This study aimed to predict potential inhibitors of Salmonella typhimurium ortho acetyl sulphydrylase synthase (StOASS), which has lower binding energy than the cocrystalized ligand pyridoxal 5 phosphate (PLP), using a computer-aided drug design approach including pharmacophore modeling, virtual screening, and in silico ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) evaluation. The screening and molecular docking of 4254 compounds obtained from the PubChem database were carried out using AutoDock vina, while a post-screening analysis was carried out using Discovery Studio. The best three hits were compounds with the PubChem IDs 118614633, 135715279, and 155773276, possessing binding affinities of −9.1, −8.9, and −8.8 kcal/mol, respectively. The in silico ADMET prediction showed that the pharmacokinetic properties of the best hits were relatively good. The optimization of the best three hits via scaffold hopping gave rise to 187 compounds, and they were docked against StOASS; this revealed that lead compound 1 had the lowest binding energy (−9.3 kcal/mol) and performed better than its parent compound 155773276. Lead compound 1, with the best binding affinity, has a hydroxyl group in its structure and a change in the core heterocycle of its parent compound to benzimidazole, and pyrimidine introduces a synergistic effect and consequently increases the binding energy. The stability of the best hit and optimized compound at the StOASS active site was determined using RMSD, RMSF, radius of gyration, and SASA plots generated from a molecular dynamics simulation. The MD simulation results were also used to monitor how the introduction of new functional groups of optimized compounds contributes to the stability of ligands at the target active site. The improved binding affinity of these compounds compared to PLP and their toxicity profile, which is predicted to be mild, highlights them as good inhibitors of StOASS, and hence, possible antimicrobial adjuvants. Full article
(This article belongs to the Special Issue New Perspectives on Chemoinformatics and Drug Design)
Show Figures

Figure 1

24 pages, 6681 KB  
Review
Leveraging Artificial Intelligence to Expedite Antibody Design and Enhance Antibody–Antigen Interactions
by Doo Nam Kim, Andrew D. McNaughton and Neeraj Kumar
Bioengineering 2024, 11(2), 185; https://doi.org/10.3390/bioengineering11020185 - 15 Feb 2024
Cited by 21 | Viewed by 7766
Abstract
This perspective sheds light on the transformative impact of recent computational advancements in the field of protein therapeutics, with a particular focus on the design and development of antibodies. Cutting-edge computational methods have revolutionized our understanding of protein–protein interactions (PPIs), enhancing the efficacy [...] Read more.
This perspective sheds light on the transformative impact of recent computational advancements in the field of protein therapeutics, with a particular focus on the design and development of antibodies. Cutting-edge computational methods have revolutionized our understanding of protein–protein interactions (PPIs), enhancing the efficacy of protein therapeutics in preclinical and clinical settings. Central to these advancements is the application of machine learning and deep learning, which offers unprecedented insights into the intricate mechanisms of PPIs and facilitates precise control over protein functions. Despite these advancements, the complex structural nuances of antibodies pose ongoing challenges in their design and optimization. Our review provides a comprehensive exploration of the latest deep learning approaches, including language models and diffusion techniques, and their role in surmounting these challenges. We also present a critical analysis of these methods, offering insights to drive further progress in this rapidly evolving field. The paper includes practical recommendations for the application of these computational techniques, supplemented with independent benchmark studies. These studies focus on key performance metrics such as accuracy and the ease of program execution, providing a valuable resource for researchers engaged in antibody design and development. Through this detailed perspective, we aim to contribute to the advancement of antibody design, equipping researchers with the tools and knowledge to navigate the complexities of this field. Full article
(This article belongs to the Special Issue Artificial Intelligence (AI) in Biomedicine)
Show Figures

Figure 1

46 pages, 25903 KB  
Review
Lysine-Specific Demethylase 1 Inhibitors: A Comprehensive Review Utilizing Computer-Aided Drug Design Technologies
by Di Han, Jiarui Lu, Baoyi Fan, Wenfeng Lu, Yiwei Xue, Meiting Wang, Taigang Liu, Shaoli Cui, Qinghe Gao, Yingchao Duan and Yongtao Xu
Molecules 2024, 29(2), 550; https://doi.org/10.3390/molecules29020550 - 22 Jan 2024
Cited by 10 | Viewed by 5093
Abstract
Lysine-specific demethylase 1 (LSD1/KDM1A) has emerged as a promising therapeutic target for treating various cancers (such as breast cancer, liver cancer, etc.) and other diseases (blood diseases, cardiovascular diseases, etc.), owing to its observed overexpression, thereby presenting significant opportunities in drug development. Since [...] Read more.
Lysine-specific demethylase 1 (LSD1/KDM1A) has emerged as a promising therapeutic target for treating various cancers (such as breast cancer, liver cancer, etc.) and other diseases (blood diseases, cardiovascular diseases, etc.), owing to its observed overexpression, thereby presenting significant opportunities in drug development. Since its discovery in 2004, extensive research has been conducted on LSD1 inhibitors, with notable contributions from computational approaches. This review systematically summarizes LSD1 inhibitors investigated through computer-aided drug design (CADD) technologies since 2010, showcasing a diverse range of chemical scaffolds, including phenelzine derivatives, tranylcypromine (abbreviated as TCP or 2-PCPA) derivatives, nitrogen-containing heterocyclic (pyridine, pyrimidine, azole, thieno[3,2-b]pyrrole, indole, quinoline and benzoxazole) derivatives, natural products (including sanguinarine, phenolic compounds and resveratrol derivatives, flavonoids and other natural products) and others (including thiourea compounds, Fenoldopam and Raloxifene, (4-cyanophenyl)glycine derivatives, propargylamine and benzohydrazide derivatives and inhibitors discovered through AI techniques). Computational techniques, such as virtual screening, molecular docking and 3D-QSAR models, have played a pivotal role in elucidating the interactions between these inhibitors and LSD1. Moreover, the integration of cutting-edge technologies such as artificial intelligence holds promise in facilitating the discovery of novel LSD1 inhibitors. The comprehensive insights presented in this review aim to provide valuable information for advancing further research on LSD1 inhibitors. Full article
Show Figures

Graphical abstract

Back to TopTop