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

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (222)

Search Parameters:
Keywords = drug discovery intelligence

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 286 KiB  
Review
Drug Repurposing and Artificial Intelligence in Multiple Sclerosis: Emerging Strategies for Precision Therapy
by Pedro Henrique Villar-Delfino, Paulo Pereira Christo and Caroline Maria Oliveira Volpe
Sclerosis 2025, 3(3), 28; https://doi.org/10.3390/sclerosis3030028 - 6 Aug 2025
Abstract
Multiple sclerosis (MS) is a chronic, immune-mediated disorder of the central nervous system (CNS) characterized by inflammation, demyelination, axonal degeneration, and gliosis. Its pathophysiology involves a complex interplay of genetic susceptibility, environmental triggers, and immune dysregulation, ultimately leading to progressive neurodegeneration and functional [...] Read more.
Multiple sclerosis (MS) is a chronic, immune-mediated disorder of the central nervous system (CNS) characterized by inflammation, demyelination, axonal degeneration, and gliosis. Its pathophysiology involves a complex interplay of genetic susceptibility, environmental triggers, and immune dysregulation, ultimately leading to progressive neurodegeneration and functional decline. Although significant advances have been made in disease-modifying therapies (DMTs), many patients continue to experience disease progression and unmet therapeutic needs. Drug repurposing—the identification of new indications for existing drugs—has emerged as a promising strategy in MS research, offering a cost-effective and time-efficient alternative to traditional drug development. Several compounds originally developed for other diseases, including immunomodulatory, anti-inflammatory, and neuroprotective agents, are currently under investigation for their efficacy in MS. Repurposed agents, such as selective sphingosine-1-phosphate (S1P) receptor modulators, kinase inhibitors, and metabolic regulators, have demonstrated potential in promoting neuroprotection, modulating immune responses, and supporting remyelination in both preclinical and clinical settings. Simultaneously, artificial intelligence (AI) is transforming drug discovery and precision medicine in MS. Machine learning and deep learning models are being employed to analyze high-dimensional biomedical data, predict drug–target interactions, streamline drug repurposing workflows, and enhance therapeutic candidate selection. By integrating multiomics and neuroimaging data, AI tools facilitate the identification of novel targets and support patient stratification for individualized treatment. This review highlights recent advances in drug repurposing and discovery for MS, with a particular emphasis on the emerging role of AI in accelerating therapeutic innovation and optimizing treatment strategies. Full article
Show Figures

Graphical abstract

40 pages, 3463 KiB  
Review
Machine Learning-Powered Smart Healthcare Systems in the Era of Big Data: Applications, Diagnostic Insights, Challenges, and Ethical Implications
by Sita Rani, Raman Kumar, B. S. Panda, Rajender Kumar, Nafaa Farhan Muften, Mayada Ahmed Abass and Jasmina Lozanović
Diagnostics 2025, 15(15), 1914; https://doi.org/10.3390/diagnostics15151914 - 30 Jul 2025
Viewed by 521
Abstract
Healthcare data rapidly increases, and patients seek customized, effective healthcare services. Big data and machine learning (ML) enabled smart healthcare systems hold revolutionary potential. Unlike previous reviews that separately address AI or big data, this work synthesizes their convergence through real-world case studies, [...] Read more.
Healthcare data rapidly increases, and patients seek customized, effective healthcare services. Big data and machine learning (ML) enabled smart healthcare systems hold revolutionary potential. Unlike previous reviews that separately address AI or big data, this work synthesizes their convergence through real-world case studies, cross-domain ML applications, and a critical discussion on ethical integration in smart diagnostics. The review focuses on the role of big data analysis and ML towards better diagnosis, improved efficiency of operations, and individualized care for patients. It explores the principal challenges of data heterogeneity, privacy, computational complexity, and advanced methods such as federated learning (FL) and edge computing. Applications in real-world settings, such as disease prediction, medical imaging, drug discovery, and remote monitoring, illustrate how ML methods, such as deep learning (DL) and natural language processing (NLP), enhance clinical decision-making. A comparison of ML models highlights their value in dealing with large and heterogeneous healthcare datasets. In addition, the use of nascent technologies such as wearables and Internet of Medical Things (IoMT) is examined for their role in supporting real-time data-driven delivery of healthcare. The paper emphasizes the pragmatic application of intelligent systems by highlighting case studies that reflect up to 95% diagnostic accuracy and cost savings. The review ends with future directions that seek to develop scalable, ethical, and interpretable AI-powered healthcare systems. It bridges the gap between ML algorithms and smart diagnostics, offering critical perspectives for clinicians, data scientists, and policymakers. Full article
(This article belongs to the Special Issue Machine-Learning-Based Disease Diagnosis and Prediction)
Show Figures

Figure 1

20 pages, 732 KiB  
Review
AI Methods Tailored to Influenza, RSV, HIV, and SARS-CoV-2: A Focused Review
by Achilleas Livieratos, George C. Kagadis, Charalambos Gogos and Karolina Akinosoglou
Pathogens 2025, 14(8), 748; https://doi.org/10.3390/pathogens14080748 - 30 Jul 2025
Viewed by 406
Abstract
Artificial intelligence (AI) techniques—ranging from hybrid mechanistic–machine learning (ML) ensembles to gradient-boosted decision trees, support-vector machines, and deep neural networks—are transforming the management of seasonal influenza, respiratory syncytial virus (RSV), human immunodeficiency virus (HIV), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Symptom-based [...] Read more.
Artificial intelligence (AI) techniques—ranging from hybrid mechanistic–machine learning (ML) ensembles to gradient-boosted decision trees, support-vector machines, and deep neural networks—are transforming the management of seasonal influenza, respiratory syncytial virus (RSV), human immunodeficiency virus (HIV), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Symptom-based triage models using eXtreme Gradient Boosting (XGBoost) and Random Forests, as well as imaging classifiers built on convolutional neural networks (CNNs), have improved diagnostic accuracy across respiratory infections. Transformer-based architectures and social media surveillance pipelines have enabled real-time monitoring of COVID-19. In HIV research, support-vector machines (SVMs), logistic regression, and deep neural network (DNN) frameworks advance viral-protein classification and drug-resistance mapping, accelerating antiviral and vaccine discovery. Despite these successes, persistent challenges remain—data heterogeneity, limited model interpretability, hallucinations in large language models (LLMs), and infrastructure gaps in low-resource settings. We recommend standardized open-access data pipelines and integration of explainable-AI methodologies to ensure safe, equitable deployment of AI-driven interventions in future viral-outbreak responses. Full article
(This article belongs to the Section Viral Pathogens)
Show Figures

Figure 1

17 pages, 2178 KiB  
Article
Enabling Early Prediction of Side Effects of Novel Lead Hypertension Drug Molecules Using Machine Learning
by Takudzwa Ndhlovu and Uche A. K. Chude-Okonkwo
Drugs Drug Candidates 2025, 4(3), 35; https://doi.org/10.3390/ddc4030035 - 29 Jul 2025
Viewed by 265
Abstract
Background: Hypertension is a serious global health issue affecting over one billion adults and leading to severe complications if left unmanaged. Despite medical advancements, only a fraction of patients effectively have their hypertension under control. Among the factors that hinder adherence to [...] Read more.
Background: Hypertension is a serious global health issue affecting over one billion adults and leading to severe complications if left unmanaged. Despite medical advancements, only a fraction of patients effectively have their hypertension under control. Among the factors that hinder adherence to hypertensive drugs are the debilitating side effects of the drugs. The lack of adherence results in poorer patient outcomes as patients opt to live with their condition, instead of having to deal with the side effects. Hence, there is a need to discover new hypertension drug molecules with better side effects to increase patient treatment options. To this end, computational methods such as artificial intelligence (AI) have become an exciting option for modern drug discovery. AI-based computational drug discovery methods generate numerous new lead antihypertensive drug molecules. However, predicting their potential side effects remains a significant challenge because of the complexity of biological interactions and limited data on these molecules. Methods: This paper presents a machine learning approach to predict the potential side effects of computationally synthesised antihypertensive drug molecules based on their molecular properties, particularly functional groups. We curated a dataset combining information from the SIDER 4.1 and ChEMBL databases, enriched with molecular descriptors (logP, PSA, HBD, HBA) using RDKit. Results: Gradient Boosting gave the most stable generalisation, with a weighted F1 of 0.80, and AUC-ROC of 0.62 on the independent test set. SHAP analysis over the cross-validation folds showed polar surface area and logP contributing the largest global impact, followed by hydrogen bond counts. Conclusions: Functional group patterns, augmented with key ADMET descriptors, offer a first-pass screen for identifying side-effect risks in AI-designed antihypertensive leads. Full article
(This article belongs to the Section In Silico Approaches in Drug Discovery)
Show Figures

Figure 1

25 pages, 4050 KiB  
Review
Network Pharmacology-Driven Sustainability: AI and Multi-Omics Synergy for Drug Discovery in Traditional Chinese Medicine
by Lifang Yang, Hanye Wang, Zhiyao Zhu, Ye Yang, Yin Xiong, Xiuming Cui and Yuan Liu
Pharmaceuticals 2025, 18(7), 1074; https://doi.org/10.3390/ph18071074 - 21 Jul 2025
Viewed by 536
Abstract
Traditional Chinese medicine (TCM), a holistic medical system rooted in dialectical theories and natural product-based therapies, has served as a cornerstone of healthcare systems for millennia. While its empirical efficacy is widely recognized, the polypharmacological mechanisms stemming from its multi-component nature remain poorly [...] Read more.
Traditional Chinese medicine (TCM), a holistic medical system rooted in dialectical theories and natural product-based therapies, has served as a cornerstone of healthcare systems for millennia. While its empirical efficacy is widely recognized, the polypharmacological mechanisms stemming from its multi-component nature remain poorly characterized. The conventional trial-and-error approaches for bioactive compound screening from herbs raise sustainability concerns, including excessive resource consumption and suboptimal temporal efficiency. The integration of artificial intelligence (AI) and multi-omics technologies with network pharmacology (NP) has emerged as a transformative methodology aligned with TCM’s inherent “multi-component, multi-target, multi-pathway” therapeutic characteristics. This convergent review provides a computational framework to decode complex bioactive compound–target–pathway networks through two synergistic strategies, (i) NP-driven dynamics interaction network modeling and (ii) AI-enhanced multi-omics data mining, thereby accelerating drug discovery and reducing experimental costs. Our analysis of 7288 publications systematically maps NP-AI–omics integration workflows for natural product screening. The proposed framework enables sustainable drug discovery through data-driven compound prioritization, systematic repurposing of herbal formulations via mechanism-based validation, and the development of evidence-based novel TCM prescriptions. This paradigm bridges empirical TCM knowledge with mechanism-driven precision medicine, offering a theoretical basis for reconciling traditional medicine with modern pharmaceutical innovation. Full article
(This article belongs to the Special Issue Sustainable Approaches and Strategies for Bioactive Natural Compounds)
Show Figures

Figure 1

20 pages, 1292 KiB  
Review
AI-Driven Polypharmacology in Small-Molecule Drug Discovery
by Mena Abdelsayed
Int. J. Mol. Sci. 2025, 26(14), 6996; https://doi.org/10.3390/ijms26146996 - 21 Jul 2025
Viewed by 545
Abstract
Polypharmacology, the rational design of small molecules that act on multiple therapeutic targets, offers a transformative approach to overcome biological redundancy, network compensation, and drug resistance. This review outlines the scientific rationale for polypharmacology, highlighting its success across oncology, neurodegeneration, metabolic disorders, and [...] Read more.
Polypharmacology, the rational design of small molecules that act on multiple therapeutic targets, offers a transformative approach to overcome biological redundancy, network compensation, and drug resistance. This review outlines the scientific rationale for polypharmacology, highlighting its success across oncology, neurodegeneration, metabolic disorders, and infectious diseases. Emphasis is placed on how polypharmacological agents can synergize therapeutic effects, reduce adverse events, and improve patient compliance compared to combination therapies. We also explore how computational methods—spanning ligand-based modeling, structure-based docking, network pharmacology, and systems biology—enable target selection and multi-target ligand prediction. Recent advances in artificial intelligence (AI), particularly deep learning, reinforcement learning, and generative models, have further accelerated the discovery and optimization of multi-target agents. These AI-driven platforms are capable of de novo design of dual and multi-target compounds, some of which have demonstrated biological efficacy in vitro. Finally, we discuss the integration of omics data, CRISPR functional screens, and pathway simulations in guiding multi-target design, as well as the challenges and limitations of current AI approaches. Looking ahead, AI-enabled polypharmacology is poised to become a cornerstone of next-generation drug discovery, with potential to deliver more effective therapies tailored to the complexity of human disease. Full article
(This article belongs to the Special Issue Techniques and Strategies in Drug Design and Discovery, 3rd Edition)
Show Figures

Figure 1

24 pages, 1349 KiB  
Review
Chemotaxonomy, an Efficient Tool for Medicinal Plant Identification: Current Trends and Limitations
by Adnan Amin and SeonJoo Park
Plants 2025, 14(14), 2234; https://doi.org/10.3390/plants14142234 - 19 Jul 2025
Viewed by 512
Abstract
This review highlights the critical role of chemotaxonomy in the identification, authentication, and discovery of bioactive compounds in medicinal plants. By analyzing secondary metabolites using techniques like UV spectroscopy, FTIR, HPLC, GC-MS, NMR, LC-MS-Qtof, and MALDI-TOF MS, chemotaxonomy ensures accurate plant identification, supporting [...] Read more.
This review highlights the critical role of chemotaxonomy in the identification, authentication, and discovery of bioactive compounds in medicinal plants. By analyzing secondary metabolites using techniques like UV spectroscopy, FTIR, HPLC, GC-MS, NMR, LC-MS-Qtof, and MALDI-TOF MS, chemotaxonomy ensures accurate plant identification, supporting the safe and effective use of plants in herbal medicine. Key secondary metabolites used in chemotaxonomic identification include alkaloids, flavonoids, terpenoids, phenolics, tannins, and plant peptides. Chemotaxonomy also facilitates the discovery of novel compounds with therapeutic potential, contributing to drug development. The integration of chemotaxonomy with genomics and proteomics allows a deeper understanding of plant biosynthesis and the mechanisms behind bioactive compound production. However, challenges due to variability in metabolite profiles and the lack of standardized methods remain, and future research should focus on developing global databases, improving standardization, and incorporating artificial intelligence and machine learning to enhance plant identification and bioactive compound discovery. The integration of chemotaxonomy with personalized medicine offers the potential to tailor plant-based therapies to individual genetic profiles, advancing targeted treatments. This review underscores chemotaxonomy’s importance in bridging traditional knowledge and modern science, offering sustainable solutions for medicinal plant use and drug development. Full article
(This article belongs to the Special Issue Plant Phylogeny, Taxonomy and Evolution)
Show Figures

Figure 1

13 pages, 1243 KiB  
Review
Evidence-Based Medicine: Past, Present, Future
by Filippos Triposkiadis and Dirk L. Brutsaert
J. Clin. Med. 2025, 14(14), 5094; https://doi.org/10.3390/jcm14145094 - 17 Jul 2025
Viewed by 627
Abstract
Early medical traditions include those of ancient Babylonia, China, Egypt, and India. The roots of modern Western medicine, however, go back to ancient Greece. During the Renaissance, physicians increasingly relied on observation and experimentation to understand the human body and develop new techniques [...] Read more.
Early medical traditions include those of ancient Babylonia, China, Egypt, and India. The roots of modern Western medicine, however, go back to ancient Greece. During the Renaissance, physicians increasingly relied on observation and experimentation to understand the human body and develop new techniques for diagnosis and treatment. The discovery of antibiotics, antiseptics, and other drugs in the 19th century accelerated the development of modern medicine, the latter being fueled further by advances in technology, research, a better understanding of the human body, and, most recently, the introduction of evidence-based medicine (EBM). The EBM model de-emphasized intuition, unsystematic clinical experience, and pathophysiologic rationale as sufficient grounds for clinical decision-making and stressed the examination of evidence from clinical research. A later EBM model additionally incorporated clinical expertise and the latest model of EBM patients’ preferences and actions. In this review article, we argue that in the era of precision medicine, major EBM principles must be based on (a) the systematic identification, analysis, and utility of big data using artificial intelligence; (b) the magnifying effect of medical interventions by means of the physician–patient interaction, the latter being guided by the physician’s expertise, intuition, and philosophical beliefs; and (c) the patient preferences, since, in healthcare under precision medicine, the patient will be a central stakeholder contributing data and actively participating in shared decision-making. Full article
(This article belongs to the Section Clinical Research Methods)
Show Figures

Figure 1

18 pages, 1790 KiB  
Article
Development of Co-Amorphous Systems for Inhalation Therapy—Part 1: From Model Prediction to Clinical Success
by Eleonore Fröhlich, Aurora Bordoni, Nila Mohsenzada, Stefan Mitsche, Hartmuth Schröttner and Sarah Zellnitz-Neugebauer
Pharmaceutics 2025, 17(7), 922; https://doi.org/10.3390/pharmaceutics17070922 - 16 Jul 2025
Viewed by 413
Abstract
Background/Objectives: The integration of machine learning (ML) and artificial intelligence (AI) has revolutionized the pharmaceutical industry by improving drug discovery, development and manufacturing processes. Based on literature data, an ML model was developed by our group to predict the formation of binary [...] Read more.
Background/Objectives: The integration of machine learning (ML) and artificial intelligence (AI) has revolutionized the pharmaceutical industry by improving drug discovery, development and manufacturing processes. Based on literature data, an ML model was developed by our group to predict the formation of binary co-amorphous systems (COAMSs) for inhalation therapy. The model’s ability to develop a dry powder formulation with the necessary properties for a predicted co-amorphous combination was evaluated. Methods: An extended experimental validation of the ML model by co-milling and X-ray diffraction analysis for 18 API-API (active pharmaceutical ingredient) combinations is presented. Additionally, one COAMS of rifampicin (RIF) and ethambutol (ETH), two first-line tuberculosis (TB) drugs are developed further for inhalation therapy. Results: The ML model has shown an accuracy of 79% in predicting suitable combinations for 35 APIs used in inhalation therapy; experimental accuracy was demonstrated to be 72%. The study confirmed the successful development of stable COAMSs of RIF-ETH either via spray-drying or co-milling. In particular, the milled COAMSs showed better aerosolization properties (higher ED and FPF with lower standard deviation). Further, RIF-ETH COAMSs show much more reproducible results in terms of drug quantity dissolved over time. Conclusions: ML has been shown to be a suitable tool to predict COAMSs that can be developed for TB treatment by inhalation to save time and cost during the experimental screening phase. Full article
(This article belongs to the Special Issue New Platform for Tuberculosis Treatment)
Show Figures

Graphical abstract

42 pages, 6065 KiB  
Review
Digital Alchemy: The Rise of Machine and Deep Learning in Small-Molecule Drug Discovery
by Abdul Manan, Eunhye Baek, Sidra Ilyas and Donghun Lee
Int. J. Mol. Sci. 2025, 26(14), 6807; https://doi.org/10.3390/ijms26146807 - 16 Jul 2025
Viewed by 1017
Abstract
This review provides a comprehensive analysis of the transformative impact of artificial intelligence (AI) and machine learning (ML) on modern drug design, specifically focusing on how these advanced computational techniques address the inherent limitations of traditional small-molecule drug design methodologies. It begins by [...] Read more.
This review provides a comprehensive analysis of the transformative impact of artificial intelligence (AI) and machine learning (ML) on modern drug design, specifically focusing on how these advanced computational techniques address the inherent limitations of traditional small-molecule drug design methodologies. It begins by outlining the historical challenges of the drug discovery pipeline, including protracted timelines, exorbitant costs, and high clinical failure rates. Subsequently, it examines the core principles of structure-based virtual screening (SBVS) and ligand-based virtual screening (LBVS), establishing the critical bottlenecks that have historically impeded efficient drug development. The central sections elucidate how cutting-edge ML and deep learning (DL) paradigms, such as generative models and reinforcement learning, are revolutionizing chemical space exploration, enhancing binding affinity prediction, improving protein flexibility modeling, and automating critical design tasks. Illustrative real-world case studies demonstrating quantifiable accelerations in discovery timelines and improved success probabilities are presented. Finally, the review critically examines prevailing challenges, including data quality, model interpretability, ethical considerations, and evolving regulatory landscapes, while offering forward-looking critical perspectives on the future trajectory of AI-driven pharmaceutical innovation. Full article
(This article belongs to the Special Issue Advances in Computer-Aided Drug Design Strategies)
Show Figures

Graphical abstract

20 pages, 1591 KiB  
Review
From Molecules to Medicines: The Role of AI-Driven Drug Discovery Against Alzheimer’s Disease and Other Neurological Disorders
by Mashael A. Alghamdi
Pharmaceuticals 2025, 18(7), 1041; https://doi.org/10.3390/ph18071041 - 14 Jul 2025
Viewed by 963
Abstract
The discovery of effective therapeutics against Alzheimer’s disease (AD) and other neurological disorders remains a significant challenge. Artificial intelligence (AI) tools are of considerable interest in modern drug discovery processes and, by exploiting machine learning (ML) algorithms and deep learning (DL) tools, as [...] Read more.
The discovery of effective therapeutics against Alzheimer’s disease (AD) and other neurological disorders remains a significant challenge. Artificial intelligence (AI) tools are of considerable interest in modern drug discovery processes and, by exploiting machine learning (ML) algorithms and deep learning (DL) tools, as well as data analytics, can expedite the identification of new drug targets and potential lead molecules. The current study was aimed at assessing the role of AI-based tools in the discovery of new drug targets against AD and other related neurodegenerative diseases and their efficacy in the discovery of new drugs against these diseases. AD represents a multifactorial neurological disease with limited therapeutics available for management and limited efficacy. The discovery of more effective medications is limited by the complicated pathophysiology of the disease, involving amyloid beta (Aβ), neurofibrillary tangles (NFTs), oxidative stress, and inflammation-induced damage in the brain. The integration of AI tools into the traditional drug discovery process against AD can help to find more effective, safe, highly potent compounds, identify new targets of the disease, and help in the optimization of lead molecules. A detailed literature review was performed to gather evidence regarding the most recent AI tools for drug discovery against AD, Parkinson’s disease (PD), multiple sclerosis (MLS), and epilepsy, focusing on biological markers, early diagnoses, and drug discovery using various databases like PubMed, Web of Science, Google Scholar, Scopus, and ScienceDirect to collect relevant literature. We evaluated the role of AI in analyzing multifaceted biological data and the properties of potential drug candidates and in streamlining the design of clinical trials. By exploring the intersection of AI and neuroscience, this review focused on providing insights into the future of AD treatment and the potential of AI to revolutionize the field of drug discovery. Our findings conclude that AI-based tools are not only cost-effective, but the success rate is extremely high compared to traditional drug discovery methods in identifying new therapeutic targets and in the screening of the majority of molecules for clinical trial purposes. Full article
Show Figures

Figure 1

12 pages, 1380 KiB  
Article
Halicin: A New Approach to Antibacterial Therapy, a Promising Avenue for the Post-Antibiotic Era
by Imane El Belghiti, Omayma Hammani, Fatima Moustaoui, Mohamed Aghrouch, Zohra Lemkhente, Fatima Boubrik and Ahmed Belmouden
Antibiotics 2025, 14(7), 698; https://doi.org/10.3390/antibiotics14070698 - 11 Jul 2025
Viewed by 736
Abstract
Background: The global spread of antibiotic-resistant bacteria presents a major public health challenge and necessitates the development of innovative antimicrobial agents. Artificial intelligence (AI)-driven drug discovery has recently enabled the repurposing of existing compounds with novel therapeutic potential. Halicin, originally developed as an [...] Read more.
Background: The global spread of antibiotic-resistant bacteria presents a major public health challenge and necessitates the development of innovative antimicrobial agents. Artificial intelligence (AI)-driven drug discovery has recently enabled the repurposing of existing compounds with novel therapeutic potential. Halicin, originally developed as an anti-diabetic molecule, has been identified through AI screening as a promising antibiotic candidate due to its broad-spectrum activity, including efficacy against multidrug-resistant pathogens. Methods: In this study, the antibacterial activity of halicin was evaluated against a range of clinically relevant multidrug-resistant bacterial strains. Bacterial isolates were first characterized using the agar disk diffusion method with a panel of 22 conventional antibiotics to confirm resistance profiles. The minimum inhibitory concentration (MIC) of halicin was then determined for selected isolates, including Escherichia coli ATCC® 25922™ and Staphylococcus aureus ATCC® 29213™, using broth microdilution according to Clinical and Laboratory Standards Institute (CLSI) guidelines. Results: Halicin demonstrated notable antibacterial activity, with MIC values of 16 μg/mL and 32 μg/mL against E. coli ATCC® 25922™ and S. aureus ATCC® 29213™, respectively. A dose-dependent inhibition of bacterial growth was observed for the majority of tested isolates, except for Pseudomonas aeruginosa, which exhibited intrinsic resistance. This lack of susceptibility is likely related to reduced outer membrane permeability, limiting the intracellular accumulation of halicin. Conclusions: Our findings support the potential of halicin as a novel antimicrobial agent for the treatment of infections caused by antibiotic-resistant bacteria. However, further investigations, including pharmacokinetic, pharmacodynamic, and toxicity studies, are essential to assess its clinical safety and therapeutic applicability. Full article
Show Figures

Figure 1

40 pages, 2828 KiB  
Review
Generative Artificial Intelligence in Healthcare: Applications, Implementation Challenges, and Future Directions
by Syed Arman Rabbani, Mohamed El-Tanani, Shrestha Sharma, Syed Salman Rabbani, Yahia El-Tanani, Rakesh Kumar and Manita Saini
BioMedInformatics 2025, 5(3), 37; https://doi.org/10.3390/biomedinformatics5030037 - 7 Jul 2025
Viewed by 2389
Abstract
Generative artificial intelligence (AI) is rapidly transforming healthcare systems since the advent of OpenAI in 2022. It encompasses a class of machine learning techniques designed to create new content and is classified into large language models (LLMs) for text generation and image-generating models [...] Read more.
Generative artificial intelligence (AI) is rapidly transforming healthcare systems since the advent of OpenAI in 2022. It encompasses a class of machine learning techniques designed to create new content and is classified into large language models (LLMs) for text generation and image-generating models for creating or enhancing visual data. These generative AI models have shown widespread applications in clinical practice and research. Such applications range from medical documentation and diagnostics to patient communication and drug discovery. These models are capable of generating text messages, answering clinical questions, interpreting CT scan and MRI images, assisting in rare diagnoses, discovering new molecules, and providing medical education and training. Early studies have indicated that generative AI models can improve efficiency, reduce administrative burdens, and enhance patient engagement, although most findings are preliminary and require rigorous validation. However, the technology also raises serious concerns around accuracy, bias, privacy, ethical use, and clinical safety. Regulatory bodies, including the FDA and EMA, are beginning to define governance frameworks, while academic institutions and healthcare organizations emphasize the need for transparency, supervision, and evidence-based implementation. Generative AI is not a replacement for medical professionals but a potential partner—augmenting decision-making, streamlining communication, and supporting personalized care. Its responsible integration into healthcare could mark a paradigm shift toward more proactive, precise, and patient-centered systems. Full article
Show Figures

Figure 1

26 pages, 2597 KiB  
Review
Strategies Used for the Discovery of New Microbial Metabolites with Antibiotic Activity
by Pablo Dasí-Delgado, Cecilia Andreu and Marcel·lí del Olmo
Molecules 2025, 30(13), 2868; https://doi.org/10.3390/molecules30132868 - 6 Jul 2025
Viewed by 672
Abstract
The discovery of new microbial metabolites is essential to combat the alarming rise in antimicrobial resistance and to meet emerging medical needs. This work critically reviews current strategies for identifying antimicrobial compounds, emphasizing the potential of microorganisms as a rich source of bioactive [...] Read more.
The discovery of new microbial metabolites is essential to combat the alarming rise in antimicrobial resistance and to meet emerging medical needs. This work critically reviews current strategies for identifying antimicrobial compounds, emphasizing the potential of microorganisms as a rich source of bioactive secondary metabolites. This review explores innovative methods, such as investigating extreme environments where adverse conditions favor the emergence of unique metabolites; developing techniques, like the iChip, to cultivate previously uncultivable bacteria; using metagenomics to analyze complex samples that are difficult to isolate; and integrates artificial intelligence to accelerate genomic mining, structural prediction, and drug discovery optimization processes. The importance of overcoming current challenges, such as replicating findings, low research investment, and the lack of adapted collection technologies, is also emphasized. Additionally, this work analyzes the crucial role of bacterial resistance and the necessity of a holistic approach involving new technologies, sustained investment, and interdisciplinary collaboration. This work emphasizes not only the current state of metabolite discovery but also the challenges that must be addressed to ensure a continuous flow of new therapeutic molecules in the coming decades. Full article
Show Figures

Figure 1

29 pages, 1320 KiB  
Systematic Review
From Lab to Clinic: How Artificial Intelligence (AI) Is Reshaping Drug Discovery Timelines and Industry Outcomes
by Doni Dermawan and Nasser Alotaiq
Pharmaceuticals 2025, 18(7), 981; https://doi.org/10.3390/ph18070981 - 30 Jun 2025
Viewed by 1603
Abstract
Background/Objectives: Artificial intelligence (AI) is transforming drug discovery and development by enhancing the speed and precision of identifying drug candidates and optimizing their efficacy. This review evaluates the application of AI in various stages of drug discovery, from hit identification to lead optimization, [...] Read more.
Background/Objectives: Artificial intelligence (AI) is transforming drug discovery and development by enhancing the speed and precision of identifying drug candidates and optimizing their efficacy. This review evaluates the application of AI in various stages of drug discovery, from hit identification to lead optimization, and its impact on clinical outcomes. The objective is to provide insights into the role of AI across therapeutic areas and assess its contributions to improving clinical trial efficiency and pharmaceutical outcomes. Methods: A systematic review followed PRISMA guidelines to analyze studies published between 2015 and 2025, focusing on AI in drug discovery and development. A comprehensive search was performed across multiple databases to identify studies employing AI techniques. The studies were categorized based on AI methods, clinical phase, and therapeutic area. The percentages of AI methods used, clinical phase stages, and the therapeutic regions were analyzed to identify trends. Results: AI methods included machine learning (ML) at 40.9%, molecular modeling and simulation (MMS) at 20.7%, and deep learning (DL) at 10.3%. Oncology accounted for the majority of studies (72.8%), followed by dermatology (5.8%) and neurology (5.2%). In clinical phases, 39.3% of studies were in the preclinical stage, 23.1% in Clinical Phase I, and 11.0% in the transitional phase. Clinical outcome reporting was observed in 45% of studies, with 97% reporting industry partnerships. Conclusions: AI significantly enhances drug discovery and development, improving drug efficacy and clinical trial outcomes. Future work should focus on expanding AI applications into underrepresented therapeutic areas and refining models to handle complex biological systems. Full article
(This article belongs to the Special Issue Artificial Intelligence-Assisted Drug Discovery)
Show Figures

Figure 1

Back to TopTop