Abstract
Globally, drug discovery and development programs are complex, multi-decade long and prohibitively expensive. Artificial intelligence (AI) and other digital health technologies have the potential to enhance and accelerate each stage of drug discovery and development, from pre-clinical target identification to post-market repurposing, and even revolutionize the entire process. Using ophthalmology as an example, this review highlights recent AI and digital health innovations in different phases of drug discovery and development. By leveraging machine learning algorithms and vast clinical and multiomics datasets, AI can rapidly identify and validate new drug targets, optimize lead compounds, and predict pharmacokinetics, pharmacodynamics and toxicity. AI-assisted multi-modal ocular biomarkers may improve treatment monitoring and support personalized medicine. Integrating AI shortens development timelines, enhances efficiency, reduces costs, and increases the success rate of new drugs. Currently, standardized regulations for AI in ocular drug development are still lacking and urgently needed to ensure safe and equitable implementation.
Subject terms: Computational biology and bioinformatics, Drug discovery, Biomarkers, Diseases, Health care, Medical research
Introduction
Drug discovery and development programs are complex, involving public-private partnerships, and typically are multi-decade long, and hugely expensive (Fig. 1). The cost of developing a new drug is estimated between 314 million and $4.4 billion1. Using ophthalmology as an example, the global ophthalmic drug market is expected to reach $66 billion by 2030, with significant investments in major sight-threatening conditions like age-related macular degeneration (AMD) and diabetic retinopathy2. Bevacizumab, originally developed for cancer, look over 20 years and billions of dollars before being “discovered” accidentally and repurposed for treatment of AMD3,4. This led to the progressive discovery and development of anti-vascular endothelial growth factor (VEGF) therapy specific for eye diseases, including AMD, diabetic retinopathy and retinal vein occlusions, which has led to considerable decline in blindness in many countries5–9. However, the process from identifying the VEGF target to obtaining US Food and Drug Administration (FDA) approval for the use of anti-VEGF agents in the eye took more than 30 years (Fig. 2). Since 2010, only 17 new drugs have gained regulatory approval from FDA for eye diseases, focusing on conditions such as AMD, open angle glaucoma, and dry eye disease (Table 1). Thus, there remains a pressing need for faster and more innovative treatment strategies for many blinding eye conditions. For instance, no current approved treatment improves visual acuity or reverses geographic atrophy (GA) in late dry AMD patients. Gene therapy (e.g., Luxturna), improves night vision but is only effective for patients with RPE65 gene mutations10. The recent UK Clinical Eye Research Strategy study (published in 2024) identified 12 main research themes, including cataract prevention, microbial keratitis treatment, community optometric care pathways, deterring refractive error progression, early detection of childhood visual disorders, and treatment of glaucoma, neurodegeneration, dry AMD, and orbital inflammatory diseases11.
Fig. 1. Phases, attrition rate, and investment in ophthalmic drug discovery and development.
This figure outlines the key stages, attrition rates, and financial investment required in the drug discovery and development process, using ophthalmology as an example. The process begins with target identification and compound screening, followed by preclinical studies assessing efficacy, toxicity, and PK/PD using in silico, in vitro, and in vivo models. Clinical development progresses through Phases I to IV, involving increasing cohort sizes to evaluate safety, dosage, efficacy, and long-term outcomes. Each stage faces significant attrition, with fewer than 1 in 10,000 compounds reaching approval. The average cost per successful drug has been estimated at $172.7 million (2000–2018). Post-market surveillance continues to assess safety and adverse events after regulatory approval. An ophthalmic case study of omidenepag isopropyl (Eybelis), a topical treatment for IOP reduction in glaucoma and ocular hypertension, exemplifies this pipeline. This structured process ensures that only the most promising and safe treatments reach patients. PK/PD pharmacokinetics/pharmacodynamics, FDA Food and Drug Administration, IOP intraocular pressure. Figure created with BioRender.com.
Fig. 2. Timeline of anti-VEGF drug discovery and development.
This figure presents the key milestones in the discovery and development of anti-VEGF therapies. Beginning with the identification of VEGF as a vascular permeability factor in 1983, the timeline traces VEGF isolation, its role in tumor angiogenesis and retinal ischemia, and the development of ranibizumab as a targeted anti-VEGF agent. Clinical trial milestones include the MARINA and ANCHOR studies in 2006 for AMD, followed by the CRUISE and BRAVO trials for RVO in 2010. FDA approval of aflibercept and ranibizumab for AMD and DME marked significant regulatory and therapeutic achievements. VPF vascular permeability factor, VEGF vascular endothelial growth factor, AMD age-related macular degeneration, RVO retinal vein occlusions, DME diabetic macular edema, FDA Food and Drug Administration. Figure created with BioRender.com.
Table 1.
Summary of FDA’s Center for Drug Evaluation and Research (CDER) approved novel drugs for eye diseases between 2010 and 2024
| Drug compound | Brand name | Mechanism of action | Disease | FDA approval | EMA approval |
|---|---|---|---|---|---|
| Perfluorohexyloctane | Miebo | Semifluorinated alkane | Dry Eye Disease | 2023 | NA |
| Lotilaner | Xdemvy | Ectoparasiticide | Demodex blepharitis | 2023 | NA |
| Avacincaptad pegol | Izervay | C5-targeted aptamer | GA secondary to AMD | 2023 | NA |
| Tebentafusp | Kimmtrak | gp100 peptide–HLA × CD3 bispecific T-cell engager | Uveal melanoma | 2022 | 2022 |
| Faricimab | Vabysmo | VEGF × ANG2 bispecific antibody | nAMD and DME | 2022 | 2022 |
| Omidenepag isopropyl | Omlonti | Prostaglandin E2 receptor agonist | OAG or OHT | 2022 | NA |
| Satralizumab | Enspryng | IL-6R-directed mAb | NMOSD | 2020 | 2021 |
| Teprotumumab | Tepezza | IGF1R-directed mAb | Thyroid eye disease | 2020 | NA |
| Brolucizumab | Beovu | VEGF inhibitor | nAMD | 2019 | 2020 |
| Cenegermin | Oxervate | Recombinant NGF | Neurotrophic keratitis | 2018 | 2017 |
| Latanoprostene bunod | Vyzulta | Prostaglandin analog | Intraocular pressure | 2017 | NA |
| Netarsudil | Rhopressa/ | RHO kinase inhibitor | OAG or OHT | 2017 | 2019 (Rhopressa) |
| Lifitegrast | Xiidra | LFA1 antagonist | Dry eye disease | 2016 | NA |
| Tafluprost | Zioptan | Selective prostaglandin F prostanoid receptor agonist | OAG or OHT | 2012 | 2008 (Taflotan) |
| Ocriplasmin | Jetrea | Recombinant truncated form of human P plasmin | Symptomatic vitreomacular adhesion | 2012 | 2013 |
| Aflibercept | Eylea | Fusion protein that binds VEGFA and PIGF | nAMD | 2011 | 2012 |
| Alcaftadine | Lastacaft | H1 histamine receptor antagonist | Allergic conjunctivitis | 2010 | NA |
Biosimilars are not included in this table. NA means this drug has not been approved by EMA.
C5 complement 5, HLA human leukocyte antigen, VEGF vascular endothelial growth factor; Ang2 Angiopoietin 2, IL-6R interlukein-6-receptor, mAb monoclonal antibody, IGF1R insulin-like growth factor 1 receptor, LFA lymphocyte function-associated antigen 1, PIGF placental growth factor, GA geographic atrophy, AMD age-related macular degeneration, nAMD neovascular age-related macular degeneration, DME diabetic macular edema, OAG open-angle glaucoma, NMOSD neuromyelitis optica spectrum disorder, FDA Food and Drug Administration, EMA European Medicines Agency.
Artificial intelligence (AI) has emerged as a promising tool to enhance and accelerate the drug discovery process. AI encompasses a range of machine learning (ML) and deep learning (DL) techniques capable of analyzing vast datasets, identifying complex patterns, and generating predictive models. These capabilities are particularly valuable in drug discovery, where AI models can optimize molecular design, predict pharmacokinetic properties, and streamline clinical trial processes. Recent advances in AI-driven drug discovery have demonstrated its ability to significantly accelerate drug development timelines (Table 2). For instance, DSP-1181, an AI-designed drug for obsessive-compulsive disorder, was developed by Sumitomo Dainippon Pharma in collaboration with Exscientia in just 12 months—far shorter than the industry average of 5 years12. Exscientia’s AI platform optimized the drug’s chemical structure, pharmacological activity, and toxicity profile, highlighting AI’s potential to expedite early-stage drug development. Halicin, developed by an AI model at MIT, is structurally distinct from conventional antibiotics and effective against drug-resistant bacteria13. By screening over 100 million molecules, AI identified novel antibacterial compounds much faster than the traditional experimental screening method14. Recent studies show that AI integration can cut early-stage development time by over 60% and increase the likelihood of clinical success, particularly in protein-based drugs, by improving the accuracy of efficacy, safety, and manufacturability predictions15. In addition, large language models (LLMs) and vision-language models (VLMs) have introduced a new level of capability, enabling AI to generate novel molecular structures, predict drug-target interactions, and analyze vast biomedical literature at an unprecedented scale16. These models not only enhance scientific research but also improve decision-making in early-stage drug development, clinical trial design, and biomarker identification17,18.
Table 2.
Summary of drugs in development that utilized AI
| Drug compound | Company | Mechanism of action | Disease | Role of AI in drug discovery |
|---|---|---|---|---|
| DSP 118112 | Exscientia & Sumitomo Dainippon Pharma | Serotonin 5 HT1A receptor agonist | Obsessive compulsive disorder | De novo molecular design |
| AI designed antibody against TL1A140 | Absci | TL1A targeted antibody | Inflammatory bowel disease | De novo molecular design |
| ISM3091141 | Insilico Medicine & Exelixis | IND ready small molecule inhibitor of anticancer target | Cancer | Target Identification & De novo molecular design |
| Small molecule KAT6A inhibitor142 | Insilico Medicine & Stemline therapeutics | Small molecule inhibitor for anticancer target | Cancer | De novo molecular design |
| INS018_055143 | Insilico Medicine | Small-molecule inhibitor of antifibrotic target TRAF2- and NCK-interacting kinase (TNIK) | Idiopathic pulmonary fibrosis | Target Identification & De novo molecular design |
| Halicin144 | MIT | Antibiotic against E coli and Acinetobacter Baumannii | Antibiotic | Drug Repurposing |
| LP-300145 | Lantern | Interacts with tyrosine kinase inhibitor (TKI) receptors via cysteine modification. | Relapsed advanced lung cancer | Patient Stratification |
| LP 184146 | Lantern | Activated via Prostaglandin Reductase 1 (PTGR1) to its cytotoxic form in cancer cells | Triple negative breast cancer | Lead Optimization and Patient Stratification |
| LP 284147 | Lantern | Damages DNA in cancer cells | Refractory lymphomas | Lead Optimization and Patient Stratification |
TL1A tumor necrosis factor-like cytokine 1A.
While several reviews have examined the role of AI in drug discovery, they often focus on individual aspects, such as molecular discovery or clinical trial optimization, without providing a comprehensive perspective19–22. Given the rapid advancement in AI and its expanding role across multiple stages of drug development, a broader synthesis is needed. In this review, we aimed to highlight how recent AI advances can facilitate/enable drug discovery and development, starting from pre-clinical target identification to clinical trials and post-trial market repurposing, using ophthalmology as a case study. Compared to general drug development, ophthalmic drug development faces unique challenges as well as opportunities such as its unique route of administration, complex physiological barriers, and potential for imaging biomarkers in characterizing and predicting responses. By addressing these challenges, AI-driven approaches have the potential to accelerate the development of novel ophthalmic therapies, improve patient outcomes, and redefine the future of precision medicine in eye care.
Discovery phase
Target identification
Many eye diseases are associated with protein misfolding and dysregulation, like AMD, cataract, corneal dystrophies, and glaucoma23. Understanding their 3D structures is crucial for researchers to comprehend their functions, mechanisms, and roles in diseases. Many drugs work by targeting specific proteins; therefore, identifying the structure of these target proteins can reveal potential binding sites, which is a critical first step in drug design. Traditionally, the gold standards for visualizing protein structures have been X-ray crystallography and/or nuclear magnetic resolution spectroscopy, and now cryo-electron microscopy24. However, these methods are extremely costly and time-consuming, and only a very small proportion of the protein structure have been discovered so far with this method25. AI-driven approaches have transformed this process by making structural predictions more efficient, and these developments align closely with Computer-Aided Drug Design (CADD), which leverages computational tools to streamline drug discovery22.
Among all AI-based algorithms developed so far, the AlphaFold series by DeepMind represents a significant breakthrough26–28. AlphaFold uses an advanced CNN to predict the 3D structures of proteins based on their amino acid sequences26. Over the past few years, AlphaFold has undergone several rounds of refinement, with each version becoming increasingly accurate, including transitioning to a transformer-based block for prediction in AlphaFold227. This advancement has profound implications for drug discovery, particularly in the field of ophthalmology. The ability to accurately predict structure and identify binding site accelerates the drug discovery process, reducing the time and cost of bringing new therapies to market. One group of Chinese researchers used AlphaFold to create 315 abnormal structural isoforms in uveal melanoma patients, providing novel insight into potential targets of treatment29. This AI-driven approach reduces the time and cost associated with experimental methods, advancing the field of CADD by incorporating protein structural data into early-stage drug design. The release of AlphaFold3 represents another leap forward in CADD, extending the prediction capabilities from protein structures to DNA, RNA, and their interactions with small molecules28. This advancement opens new avenues for nucleic acid-based therapies in ophthalmology, where understanding complex molecular interactions is critical for treating conditions such as retinal degeneration or inherited genetic disorders. AlphaFold3’s ability to predict molecular interactions enhances ligand-based design, another core aspect of CADD, by providing detailed structural information on potential drug binding sites.
In addition, AI-driven multi-omics analysis has emerged as another powerful strategy for therapeutic target identification. PandaOmics, an AI-powered platform, integrates gene expression, proteomics, and text-based knowledge graphs to systematically identify and rank potential drug targets30. By analyzing diverse molecular datasets, PandaOmics enhances target discovery beyond traditional protein-structure approaches, prioritizing novel, druggable genes for further investigation. Expanding on this concept, AI has also been leveraged to explore non-traditional biological datasets for drug discovery. For instance, one research group employed a multitask deep learning framework to analyze the proteomes of extinct organisms (the “extinctome”), systematically identifying novel antibiotic peptides with therapeutic potential31,32 Similar AI-driven methodologies have also been applied to the human microbiome, enabling the discovery of antimicrobial peptides with promising therapeutic applications33,34. These innovative AI-based strategies in antibiotic discovery highlight a broader shift toward computationally guided drug development, which could similarly transform ocular drug discovery by identifying peptide-based treatments for ocular infections and addressing antimicrobial resistance, a recognized global health threat14,35.
Computer-aided drug design
After identifying promising targets, the next crucial step is to simulate drug interactions and optimize potential candidates. Structure-based drug design (SBDD) and ligand-based drug design (LBDD) represent two fundamental approaches in CADD. SBDD leverages the 3D structure of biological targets to identify drug candidates. It employs molecular docking to predict the optimal binding orientation and affinity of small molecules, while molecular dynamics (MD) simulations refine these predictions by modeling protein-ligand interactions over time, accounting for flexibility and conformational changes. LBDD, in contrast, focuses on known active compounds rather than target structures. A key method, quantitative structure-activity relationship (QSAR) modeling, uses statistical and machine learning approaches to predict drug efficacy based on molecular features, guiding candidate selection even when structural data is unavailable.
Molecular docking facilitates the examination of the interaction between small molecules and their target proteins. AI-powered docking approaches have revolutionized this process by predicting the binding affinity and orientation of millions of potential drug molecules more quickly and efficiently than traditional methods, as it uses DL models to predict docking outcomes for large chemical libraries36,37. For example, Deep Docking can quickly filter and prioritize molecules that may effectively bind to vascular endothelial growth factor (VEGF), a key target in AMD, accelerating the drug discovery process. An exciting example is the development of INS018_055, a TRAF2- and NCK-interacting kinas (TNIK) inhibitor for idiopathic pulmonary fibrosis (IPF), completed in just 18 months using PandaOmics and advanced molecular docking38. This AI-driven workflow combined pathway modeling, single-cell perturbation analysis, and de novo molecular docking, refining TNIK’s role in fibrosis. AI-assisted docking simulations further optimized INS018_055’s drug-likeness and stability, significantly expediting lead optimization. The inhibitor demonstrated strong anti-fibrotic and anti-inflammatory effects in preclinical models and rapidly progressed to a phase 1 clinical trial, highlighting the value of AI in target identification and early drug development.
MD simulations help researchers understand how proteins/peptides and drugs interact at an atomistic level in simulated dynamic biological environments39,40. For instance, in ocular drug discovery, MD simulations can model how a potential drug designed for the retina behaves within the aqueous humor, providing insights into the drug’s binding stability and efficacy. This is especially useful for treating complex ocular conditions such as glaucoma and AMD, where drug stability and sustained efficacy are crucial. MD simulations have shown potential in guiding the design of antimicrobial peptides (or host defense peptides), revealing the secondary structures, dissecting the mechanism of action, and identifying key amino acid residues responsible for their efficacy and toxicity35,41. For instance, Ting et al. leveraged the atomistic insights gained from MD simulations to optimize the sequence/composition of synthetic host defense peptides, which demonstrated potent in vivo efficacy and safety against drug-resistant bacterial keratitis41,42.
QSAR models play a central role in this approach by leveraging molecular descriptors to predict drug-target interactions, facilitating rational drug design and lead optimization43. Building upon traditional QSAR, Redshaw et al. recently developed highly accurate peptide-based ML models, which could predict the structure-activity relationship of antimicrobial peptides that exhibit potent in vivo activity against Staphylococcus aureus bacterial keratitis35. This suggests that ML-based QSAR approach may accelerate the discovery of new antimicrobial therapy for treating ocular infections. DL-based QSAR (DeepQSAR) integrates DL techniques such as neural networks to enhance molecular representation and prediction accuracy43. For instance, generative AI (GenAI) models like SyntheMol have been developed to design novel, structurally diverse peptide-based therapeutics44. By leveraging a Monte Carlo tree search algorithm, SyntheMol efficiently explores vast chemical spaces to generate antimicrobial candidates that are both novel and synthesizable. Combined approach utilizing deep generative models and high-throughput MD simulations has also shown promise in accelerating antimicrobial drug discovery45. This strategy demonstrated remarkable speed, with two potent and low-toxicity antimicrobial peptides discovered, synthesized, and validated within just 48 days—significantly faster than conventional drug development timelines45. All these approaches could be adapted to enable the discovery of novel antimicrobial therapies for tackling ocular infections and antimicrobial resistance14. Beyond innovation, accessibility and cost-effectiveness are also key considerations in antimicrobial drug development. ZairaChem automated pipeline combines various ML models to conduct QSAR and quantitative structure-property relationship analyses on chemical libraries46. This approach allows for efficient prediction of drug efficacy and safety profiles, even in resource-limited settings like those in Africa.
Explainable AI (XAI) is another crucial tool for molecular discovery, as it offers transparency and interpretability in AI-driven predictions. Ocular diseases such as AMD and glaucoma involve complex molecular pathways, and XAI can enhance early-stage drug discovery by elucidating the structural and functional relationships between target proteins and small molecules. One significant advantage of XAI lies in feature attribution methods, such as Shapley Additive Explanations (SHAP) and Local Interpretable Model-agnostic Explanations (LIME), which help identify key molecular descriptors contributing to drug-target interactions47. This is particularly useful in understanding why certain ligand-protein interactions are favorable, providing insights into optimal drug candidates while mitigating biases inherent in black-box models48. One example of XAI in drug development is SmartCADD, a platform that integrates AI and quantum mechanics principles for virtual screening, improving the interpretability of docking scores and ligand binding affinity predictions49. Unlike traditional docking approaches, SmartCADD employs a modular, open-source framework that combines DL-based virtual screening with quantum-informed scoring functions, allowing for more precise predictions of ligand binding affinity and ADMET (absorption, distribution, metabolism, excretion, and toxicity) properties. By incorporating explainability tools such as SubgraphX, SmartCADD enhances interpretability by visualizing key molecular substructures that influence binding interactions, facilitating more informed decision-making in drug optimization.
More recently, the advent of reasoning-based models like OpenAI o1, Google Gemini, and DeepSeek R1 represents another leap in AI’s capability for complex problem-solving50–52. While these models do not yet have specific applications in drug discovery, their potential to reason through complex biochemical interactions, optimize multi-objective problems, and generate novel hypotheses makes them an exciting frontier. Future developments may enable these models to rationalize AI-generated predictions in drug discovery, interpret omics data more effectively, or even assist in multi-modal drug design by integrating molecular docking, systems biology, and clinical datasets into a cohesive framework.
In vitro ADMET simulations
Once the molecular target has been established, extensive rounds of preclinical experiments and simulations are usually required to better understand the dose-response and ADMET profile before progressing to clinical trials. Traditionally, there has been a significant reliance on animal models; however, these models come with their own set of challenges, including ethical concerns, issues with cross-species extrapolation and thus high failure rates, and high costs53.
In this context, organoids and organ-on-a-chip (OoC) technologies have emerged as promising alternatives to animal models53. Organoids are three-dimensional structures grown from stem cells that mimic the architecture and function of real organs54. OoC devices, on the other hand, are microfluidic platforms that simulate the physiological responses of entire organs. Both technologies offer more physiologically relevant systems for studying drug effects, mechanisms of action, and potential toxicity, potentially reducing reliance on animal models. While organoids and OoC technologies may not completely replace animal testing, they can help limit the number of animal groups required by offering quicker, more cost-effective, and human-relevant data. This can enhance the predictive accuracy of human drug responses, particularly in early preclinical stages.
Several OoC models have been proposed to study the retina and ocular surface, providing valuable insights into ocular drug development. A retina-on-a-chip model integrates human iPSC-derived retinal organoids (ROs) with vascular-like perfusion, allowing for the first time the recapitulation of mature photoreceptor-RPE interactions, critical for modeling retinal toxicity and drug screening55. Another innovative model, the Wet-AMD-on-a-chip, mimics the outer blood-retinal barrier and allows researchers to study choroidal neovascularization in a physiologically relevant manner, which is essential for testing anti-VEGF therapies56. Additionally, a biomimetic artificial eye model has successfully reconstructed the ocular surface, complete with blinking and tear film dynamics, enabling disease modeling for dry eye disease and high-content drug screening57. These models represent significant strides in ophthalmic research by bridging the gap between in vitro and in vivo systems, improving drug discovery, toxicity assessment, and therapeutic development.
The integration of AI with OoC can significantly further enhance the potential of existing technology. One of the primary benefits of AI in OoC technology is its ability to handle and analyze the large datasets generated by these systems. DL models can process data from various sensors and imaging modalities, providing detailed insights into cellular responses and drug efficacy. This is particularly beneficial for studying complex ocular diseases, where traditional methods struggle to capture the intricate ocular microenvironment. For instance, AI has been successfully applied to angiogenesis, an essential component of diabetic retinopathy, in OoCs by using DL for vessel segmentation and quantification58. Additionally, AI also supports the automation of experimental procedures in OoCs, enhancing reproducibility and throughput59. Recent advancements have enabled reinforcement learning algorithms to autonomously optimize microfluidic conditions, dynamically adjusting flow rates and culture environments to enhance cell viability and mimic physiological conditions59. An automated microfluidic platform has already demonstrated the ability to precisely control drug dosing and sequencing for improved treatment outcomes in pancreatic tumor organoids60. Recent advances have also demonstrated the emerging role of AI in enabling phenotype-based, high-throughput screening in 3D organoid systems. For example, AI-driven interferometry and machine learning-based segmentation allow for dynamic, label-free quantification of treatment responses in thousands of individual organoids at single-organoid resolution61. Similarly, 3D convolutional neural networks such as Stardist-3D have enabled cellular-resolution phenotyping of retinal organoids by distinguishing subclonal responses and spatial architecture62. These approaches go beyond traditional endpoints to capture subtle, heterogeneous drug effects in complex tissues. Applying similar AI-integrated approaches to retinal or ocular organoids could significantly improve the throughput and precision of ophthalmic drug discovery, by enabling phenotype-based screening and optimizing microfluidic flow for intraocular drug delivery and disease modeling—ultimately allowing for more precise, functionally relevant, and effective treatments.
Physiologically-based pharmacokinetics (PBPK) modeling is another valuable tool in preclinical studies. It predicts the ADMET of drugs in the body. PBPK models are comprised of complex parametric mathematical models that simulate the behavior of drugs within various biological compartments63,64. Accurate physiological and biochemical parameters are crucial for these models to make precise predictions, but obtaining such parameters can be challenging due to biological variability and limitations in experimental data. The sophisticated physiological structures, distinctive barriers, complicated biomechanical processes, and diverse delivery routes make deriving such parameters even more difficult for the eye65. For instance, the ocular surface presents several physiological barriers, such as the tear film, blinking, tear drainage, and structural barriers such as corneal and conjunctival epithelium. Additionally, the choice of drug delivery routes—including intravitreal, subconjunctival, and intracameral injections—can significantly influence a drug’s ADME profile and therapeutic outcomes66. As such, animal models are often required to validate the findings observed in in vitro settings67.
To address these challenges and reduce reliance on animal testing, AI offers a powerful alternative for simulating intricate ocular pharmacokinetic processes. For example, it can model corneal permeability by integrating drug physicochemical properties and formulation data, predict intraocular distribution across compartments like the vitreous humor and posterior segment using imaging and anatomical datasets, and anticipate ocular metabolism by identifying relevant enzymatic pathways and turnover rates. These capabilities are especially valuable for optimizing drug delivery strategies, improving ocular bioavailability, and enabling the development of next-generation ophthalmic therapeutics. This can be achieved through several key mechanisms. Firstly, AI algorithms have been widely used for data mining, enabling them to derive key physiological and biochemical parameters more precisely than traditional methods68. By ensuring more reliable parameter estimates, AI contributes to more accurate simulations and predictions, facilitating better drug development decisions. Secondly, AI contributes to the development of innovative ML-based, nonparametric methods68. Unlike traditional parametric models that are limited by fixed assumptions, nonparametric AI models can adapt to new data, learning complex patterns and relationships that may not be immediately apparent. This flexibility allows them to provide more accurate predictions for novel drugs, particularly those with unique or poorly understood properties. A real-life worked example is the application of Neural-ODE models in predicting treatment regimen for trastuzumab emtansine, a drug for breast cancer, and research efforts based on novel structures like long short-term memory networks are undergoing69,70. Different areas of AI applications in drug discovery are summarized in Fig. 3.
Fig. 3. Artificial intelligence in molecular drug discovery.
This figure summarizes how AI accelerates the molecular discovery process across several key steps. Beginning with target identification, AI integrates peptide sequences, multi-omics data, and molecular structure prediction to prioritize potential drug targets. CADD further enables structure-based and ligand-based drug design to develop and refine drug prototypes. AI also enhances in vitro simulation through advanced platforms like organoids, organ-on-a-chip systems, and PBPK modeling. These simulations inform early ADMET analysis, improving the prediction of a compound’s behavior in humans. AI artificial intelligence, CADD computer-assisted drug design, ADMET absorption, distribution, metabolism, excretion, and toxicity, PBPK physiologically-based pharmacokinetics. Figure created with BioRender.com.
Development phase
Biomarkers for response monitoring
Recent advancements in high-throughput sequencing and single-cell analysis have significantly enhanced our understanding of the molecular impacts of drugs, providing a robust foundation for targeted therapeutic interventions. The depth of molecular insights gained, particularly concerning the physiological effects of drugs, allows for dynamic and adaptive enrollment strategies in clinical trials, potentially maximizing success rates. For instance, whole genome sequencing can be employed to stratify patients based on their genotypic variations, enabling more personalized medicine approaches and improving treatment efficacies71. Unlike conventional pharmacogenomics, which focuses on single nucleotide polymorphisms, AI facilitates network pharmacology by investigating the complex dynamic networks of gene-gene and gene-environment interactions that affect drug responses72. Moreover, substantial efforts have been directed toward studying drug-omics associations, which can effectively decode the intricate interactions between drugs and biomolecules73.
The eye presents unique opportunities for leveraging such technologies due to its highly specific molecular biomarkers, including proteins, DNA, and metabolites from tears, aqueous humor, and vitreous humor. In contrast to systemic biomarkers, ocular biomarkers provide a more precise understanding of the ocular microenvironment since they are locally enriched. These biomarkers can be accessed through liquid biopsies, a method that has proven to be not only insightful for understanding underlying disease mechanisms but also safe for patients74. Liquid biopsy techniques in ophthalmology allow for the detection and analysis of these biomarkers with minimally invasive procedures, offering a real-time snapshot of the molecular landscape within the eye75. This approach enhances the ability to monitor disease progression and response to treatment making it an indispensable tool in developing new therapeutic strategies. For instance, various complement factors from aqueous humor are closely associated with the pathophysiology of AMD and glaucoma, thus serving as potential treatment targets76.
Furthermore, improvements in computational power have enabled a systems biology approach that encompasses various “omic” biomarkers, including epigenomic, transcriptomic, proteomic, lipidomic, and metabolomic data, thereby providing a more holistic perspective on pathophysiology. Researchers have constructed transcriptome and proteomic networks for both healthy and diseased eyes (e.g. diabetic retinopathy) to create a more comprehensive understanding of these conditions77,78. With AI, it is now possible to identify cellular mechanisms underlying eye aging, which could pave the way for novel therapeutic interventions79. Proteomic biomarkers also are being used to analyze responses to treatments for geographic atrophy80. However, a key challenge in leveraging omics data is mitigating confounding factors that obscure true biological signals. CODE-AE (context-aware deconfounding autoencoder) addresses this by disentangling intrinsic biomarkers from technical and biological noise, such as batch effects or patient variability, through a specialized self-supervised learning framework81. By learning shared biological patterns while suppressing dataset-specific distortions, CODE-AE improves model robustness in predicting patient-specific drug responses. Applying similar AI-driven strategies to ophthalmology could enhance precision medicine approaches for ocular diseases, improving biomarker-based diagnostics and therapeutic targeting. As our understanding of the molecular basis of eye diseases expands, AI-driven approaches are expected to play a crucial role in the discovery and development of new ophthalmic drugs by incorporating multi-omics and genomics to better predict individual outcomes and responses, ultimately improving patient outcomes and advancing the field of ocular therapeutics.
In the domain of ocular diagnostics, imaging technologies hold an even more significant place than molecular assays. Technologies such as color fundus photography, fluorescein angiography, optical coherence tomography (OCT), and OCT angiography are imperative for detailed ocular examination, and next-generation investigational modalities leveraging visible light OCT, fluorescence and multispectral imaging, and other new technologies can gather even deeper datasets on ocular and particularly retinal structure, function, metabolism, and physiology82–84. AI algorithms enhance the capabilities of these imaging methods by providing fast and accurate analysis of key features associated with diseases, such as vasculature and layer thickness85,86. This level of quantification is both highly reliable and precise, reducing the reliance on human graders, saving time, and speeding up diagnostic processes87. Recently, unsupervised learning and advent of vision-language model has enabled AI to discover similar patterns among diseased patients without specific pre-defined labels or domain expertise88. This ability brings the understanding of disease morphology to a higher level, as the algorithm can detect nuanced changes that humans cannot see, such as those in AMD, sarcoidosis and even some rare retinal conditions88–90.
Similarly, AI can be utilized to quantify structural changes in response to treatment and detect subclinical changes that may not be visible at first91,92. Therefore, AI algorithms can predict patient’s response to treatment and determine treatment frequency based on the biomarkers extracted from the images93,94. This not only helps identify eligible patients for treatment but also maximizes treatment success rate through a more personalized, effective treatment regimen. GenAI is able to synthesize hypothetical post-treatment images, helping clinicians and patients visualize treatment responses more intuitively95. Moreover, AI-detected changes may serve as secondary endpoints during trials, potentially providing go/no-go decision-making support in early phase trials, speeding up the drug approval process and bringing innovations to patients sooner96,97. For example, in glaucoma, where understanding and preserving neural structures is as important as managing intraocular pressure (IOP), AI can analyze changes in the thickness of the retinal nerve fiber layer, moving beyond the traditional focus on IOP reduction. Newer therapeutic avenues towards neuroprotection, for example in optic neuropathies, may fully rely on such AI-derived insights from novel modalities to measure meaningful outcomes in retinal or optic nerve physiology98,99. This introduces a more comprehensive approach to understanding treatment response. Currently, AI has been used in various secondary analyses of existing clinical trial data and has proven its values (Table 3).
Table 3.
A selected summary of the current studies using deep learning for secondary analysis of clinical trial data or design of clinical trials in ophthalmology
| Author name (year) | Study population | Sample size | Treatment | Condition | Imaging | Outcome of interest |
|---|---|---|---|---|---|---|
| Quantify structural changes | ||||||
| Schmidt-Erfurth et al.148 | HAWK and HARRIER | 1078 and 739 | Brolucizumab OR Aflibercept | nAMD | OCT | IRF, SRF, PED |
| Fu et al.149 | FILLY | 197 | Pegcetacoplan | GA | OCT | GA area, RPE loss, hypertransmission, PR degeneration, and intact macular area |
| Vogl et al.150 | FILLY | 312 | Pegcetacoplan | GA | OCT | LPR, PR thickness, HRF concentration |
| Roberts et al.151 | DRCR Network | 570 | Aflibercept, Ranibizumab, OR Bevacizumab | DME | OCT | IRF, SRF |
| Schmidt-Erfurth et al.152 | OAKS and DERBY | 897 | Pegcetacoplan | GA | OCT | RPE loss and PR degeneration |
| Predicting functional restoration | ||||||
| Mulyukov et al.153 | HAWK and HARRIER | 594 + 389 | Brolucizumab OR Aflibercept | nAMD | OCT | Disease activity score |
| Chandra et al.154 | CATT | 1029 | Ranibizumab OR Bevacizumab | nAMD | OCT | BCVA |
| Kikuchi et al.155 | AVENUE | 273 | Faricimab | nAMD | OCT | BCVA and CST |
| Maunz et al.156 | HARBOR | 432 | Ranibizumab | nAMD | OCT | BCVA |
| Predicting treatment frequency | ||||||
| Bogunovic ´ et al.157 | HARBOR | 317 | Ranibizumab | nAMD | OCT | Low (≤5 injection), High (≥16) |
| Chandra et al. 158 | CATT | 493 | Ranibizumab OR Bevacizumab | nAMD | OCT | Low (≤8), High (≥19) |
nAMD neovascular age-related macular degeneration, GA geographical atrophy, DME diabetic macular edema, OCT optical coherence tomography, IRF intraretinal fluid, SRF subretinal fluid, PED pigment epithelial detachment, RPE retinal pigment epithelium, PR photoreceptor, LPR local progression rate, HRF hyperreflective foci, BCVA best corrected visual acuity, CST central subfield thickness.
Clinical trial implementation
One of the key challenges in clinical trials has been suboptimal patient cohort selection, inefficient recruiting techniques, and difficulties in maintaining effective monitoring throughout the trial period. These challenges are major contributors to high trial failure rates. Traditional methods often rely on electronic health records (EHR) and manual processes to identify eligible participants, which can miss key patients or include those who do not meet all criteria. AI-driven predictive analytics and ML algorithms address these issues by refining patient selection and recruitment strategies, a process known as clinical trial enrichment20. By analyzing large datasets, including medical histories, laboratory investigations, and even lifestyle factors, AI can identify the most appropriate candidates for specific trials with greater accuracy. For instance, AI algorithms based on OCT scans have demonstrated higher precision in identifying eligible patients for AMD clinical trials compared to conventional EHR systems alone, improving the representativeness and appropriateness of the patient cohort100.
Moreover, AI facilitates the operation of decentralized clinical trials, which allow participants to engage in the study from various locations, not limited to a few centralized trial sites101. This approach significantly expands the participant pool, particularly for diseases like AMD and diabetic retinopathy, which predominantly affect elderly populations who may have mobility constraints. Furthermore, AI enables cross-center collaboration by allowing multiple trial sites to share insights and data in real-time through techniques like federated learning. Federated learning supports the analysis of aggregated data from different trial sites without compromising patient privacy, as individual data remains localized102. This helps address data privacy concerns while also improving the robustness of trial outcomes by drawing on a larger, more diverse dataset.
Adaptive trial designs supported by AI further enhance the efficiency of clinical trials. In traditional trials, protocols are often static and predetermined, which can lead to delays or resource waste if the initial hypotheses prove ineffective. AI-driven adaptive trials dynamically adjust protocols based on interim results, allowing researchers to focus resources on the most promising therapeutic avenues while discontinuing less effective treatments earlier in the process103. These platforms also streamline trial management by automating tasks such as data entry, and adverse event reporting, and patient follow-up, increasing the overall efficiency and accuracy of trials. This not only speeds up the trial but also reduces the financial burden associated with lengthy and ineffective studies. There have already been studies incorporating AI into clinical design in real life. Real-world examples of AI integration in clinical design are already emerging. For instance, the Vibe Up study is a decentralized AI-adaptive group sequential randomized controlled trial that compares the effectiveness of digital self-guided interventions in reducing self-reported psychological distress104. Unlike traditional randomized controlled trials (RCTs), which require fixed treatment allocations, Vibe Up employs a bandit-based response adaptive trial method, meaning that as new data emerges, the AI system dynamically adjusts participant allocation to prioritize the most effective interventions. This AI-driven approach not only reduces sample size requirements but also improves efficiency by optimizing treatment allocation in real time, demonstrating how adaptive methodologies can make clinical trials more responsive and cost-effective.
Additionally, the integration of AI-generated Digital Twins into clinical trials is revolutionary. Digital Twins are AI-driven virtual representations of trial participants, constructed from real-world clinical data, including biometrics, imaging, genomics, and molecular markers105. Digital Twins are revolutionizing clinical trial efficiency by reducing the reliance on large control groups, enabling adaptive trial designs, and predicting treatment responses with greater accuracy106. Traditionally, RCTs require large control arms, where participants receive a placebo or standard-of-care treatment. However, AI-powered Digital Twins can generate synthetic control arms—statistical representations of placebo groups derived from historical clinical trial data—thereby reducing the number of real patients needed in these groups. This minimizes ethical concerns associated with withholding treatment from participants and accelerates trial timelines. Additionally, Digital Twins enable prognostic covariate adjustment (PROCOVA), an AI-driven statistical method that improves trial efficiency by enhancing statistical power105. This technique has been qualified by the European Medicines Agency (EMA) and aligns with existing FDA guidelines, allowing for smaller, more efficient trials while maintaining rigorous scientific standards105. Furthermore, AI-driven Digital Twins continuously update with new biomarker data, refining predictions of individual treatment responses. In ophthalmology, for example, Digital Twins could integrate multi-modal imaging, molecular biomarkers, and AI-driven risk models to stratify patients based on their likelihood of responding to treatment. This enables adaptive trial designs, where treatment regimens are dynamically adjusted based on real-time patient response data106. In ophthalmology, Digital Twin applications are beginning to take shape across multiple disease areas. For instance, in proliferative vitreoretinopathy, Digital Twins have been proposed to model the disease’s complex fibrotic pathways and simulate responses to anti-fibrotic agents such as Nintedanib107. Similarly, in glaucoma, Digital Twins can incorporate multimodal clinical data—including intraocular pressure, optic nerve imaging, and genetic risk factors—to forecast disease progression and support personalized treatment planning108. By integrating patient-specific molecular and imaging data, these models can predict therapeutic efficacy and help optimize treatment regimens without the need for extensive in vivo testing.
The integration of DL algorithms with smartphone technology and wearable devices further simplifies the trial process by making monitoring more accessible and user-friendly109,110. Smartphones equipped with AI-based diagnostic tools can facilitate regular ocular health monitoring by patients themselves, enabling continuous data collection throughout the trial. This not only empowers patients to take a more active role in their healthcare but also provides real-time data to pharmaceutical companies and researchers. The ability to monitor drug performance more closely allows for timely adjustments to treatment protocols, thereby improving the overall effectiveness of clinical trials. For instance, a study conducted in China demonstrated that patients managed by a combination of primary care providers and LLMs had better self-management behavior and adherence to follow-up care for diabetic retinopathy treatment compared to those managed by primary care providers alone111. This improvement is likely due to LLM’s ability to provide more personalized and data-driven recommendations, while also offering structured guidance that enhances communication and decision-making between patients and providers. Additionally, LLM-generated responses may improve patient engagement by increasing trust and accessibility of medical information. Such advancements highlight the expanding role of AI in enhancing diagnostic accuracy and treatment efficacy in ophthalmology, paving the way for more personalized and effective healthcare solutions.
Post-market drug repurposing
Drug development is indeed a time-consuming and expensive process. In this context, the repurposing of existing, proven-safe drugs for new therapeutic uses offers a compelling alternative112. This approach not only saves time and reduces costs significantly but also minimizes risk, as the safety profiles of these drugs are already well-understood. Additionally, drug repurposing can expedite the availability of treatments, particularly in response to urgent health crises where new drug discovery and approval might be too slow. It also extends the commercial life of drugs, providing pharmaceutical companies with new markets and patients with more treatment options. In today’s era of big data and extensive data sharing, the use of AI in drug repurposing is increasingly viable and valuable. AI can integrate and analyze diverse data types from global datasets, offering insights that might not be apparent through traditional research methods. This technological approach is becoming an integral part of the pharmaceutical industry’s strategy to enhance drug development efficiency and responsiveness to emerging health challenges.
Natural language processing and DL are pivotal in identifying repurposing opportunities by extrapolating from vast amounts of existing data, including published research, clinical trial data, and EHR. Various studies confirmed its feasibility in conditions like coronary artery disease, Alzheimer’s disease, and Parkinson’s disease, and such capability was most notably demonstrated during the COVID-19 pandemic113,114. Remdesivir, an anti-viral drug initially designed for the Ebola virus, was identified by a DL model as one of the potential treatments for COVID-19114. Subsequent clinical trials also confirmed its benefits, leading to emergency use authorization by regulatory bodies worldwide115. This real-life example showcased how AI could leverage big data to identify potential drug repurposing candidates effectively. Recently, advances in GenAI, particularly LLMs like ChatGPT, have pushed the limits of AI in drug repurposing to new heights18,116,117. GenAI excels at processing unstructured data and can identify non-obvious connections, making it an invaluable tool in drug discovery and repurposing. Despite its recent emergence, researchers have already applied GenAI to identify potential drug-repurposing candidates for Alzheimer’s disease118. Although there have not yet been eye-specific applications, it is only a matter of time before GenAI is utilized in ophthalmic research and drug discovery.
In addition to leveraging real-world efficacy data from EHRs, repurposing can also be guided by AI-powered omics-based approaches. Recent studies have demonstrated that combined multi-omics with AI-driven computational modeling can enhance drug discovery and repositioning. For instance, AI-based omics analysis has been applied to identify drug pairs for pyroptosis therapy in triple-negative breast cancer, integrating transcriptomic data, drug databases, and biofactor-regulated neural networks119. Similar methodologies could be employed in ophthalmology, identifying systemic drugs with beneficial effects on eye diseases. Figure 4 summarizes the AI applications throughout the entire phase of drug discovery and development.
Fig. 4. Applications of artificial intelligence and digital health across the drug development pipeline.
In the discovery phase, AI supports target identification through structural prediction and multi-omics integration, and enhances computer-aided drug design via molecular modeling and docking. ADMET simulation benefits from organoids, organ-on-a-chip systems, and PBPK modeling. During development, biomarker monitoring incorporates ocular imaging (e.g., CFP, OCT) and fluid-based biomarkers (aqueous and vitreous humor) for evaluating treatment response. Trial implementation is advanced by decentralized designs, digital twins, and wearable technologies. Finally, drug repurposing leverages AI using EHR data and molecular analytics to identify new uses for existing therapies. ADMET absorption, distribution, metabolism, excretion, and toxicity, PBPK physiologically-based pharmacokinetics, CFP colored fundus photography, OCT optical coherence tomography, EHR electronic health records. Figure created with BioRender.com.
Limitations
Regulatory considerations
While AI holds great potential in revolutionizing drug discovery, it also comes with limitations. Given the extensive data requirements for training AI systems, concerns about data privacy and security have emerged120. Data sharing across institutions, countries or industries often leads to exposure risks of sensitive health data. Regulations such as the General Data Protection Regulation and the Health Insurance Portability and Accountability Act should be adhered to protect patient confidentiality and privacy. A promising solution to address these concerns is federated learning, a technique which enables raw data to remain with its source, thereby minimizing the risk of exposure. Clients train local ML models on their own data, sending only updated model parameters to a central server for aggregation121. The server then shares aggregated parameters back with clients for further training. This process repeats until the model achieves the desired accuracy121. By facilitating collaboration between AI models, federated learning can enhance data privacy while contributing to the development of more accurate ML models in drug discovery, such as Alphafold and complementary technologies such as OoC.
The United States Food and Drug Administration’s (FDA) has led efforts to regulate AI in drug discovery, given its implications on treatment and prognosis. The FDA has reviewed hundreds of regulatory submissions involving AI in drug development. In March 2024, it introduced a risk-based regulatory framework outlining four areas of focus: (1) fostering collaboration to safeguard public health; (2) promoting development of harmonized standards, guidelines, best practices, and tools; (3) advancing the development of regulatory approaches that support innovation; and (4) supporting research related to evaluation and monitoring of AI performance122. Globally, the EMA and the Medicines and Healthcare products Regulatory Agency (MHRA) are developing standardized protocols for AI and ML in drug discovery. Regulatory frameworks struggle to keep pace with AI’s rapid evolution. Current drug discovery regulations lack provisions for GenAI and LLMs, which tend to produce unpredictable outputs and challenges123. These regulations should extend to AI and bioinformatics for analysis of aqueous or vitreous humor biomarkers to uphold ethical standards in AI usage in drug development. Currently, there lacks a unified international guideline. A coordinated and global effort for more consistent and united regulations for applying AI in drug discovery are needed124. Additionally, a life cycle management approach—monitoring AI models post-market—could improve implementation but remains difficult to enforce123.
AI’s role in drug discovery raises complex patentability and intellectual property concerns. The United States Patent and Trademark Office allows patents for “any new and useful process”, but algorithms, as abstract procedures, may not qualify125. This is further complicated by the Patient Eligibility Restoration Act of 2023 which excludes mental processes and mathematical formulas from eligibility125. Concerns regarding the originality of AI-aided or AI-generated therapeutics have been raised. Under the Federal Circuit, an inventor must be a natural person, hence an AI algorithm cannot be a named inventor126. Enablement and written description requirements in section 112 have been hinderances, especially when vague descriptions of “ML” and “neural networks” have been used without specific details of the method. To avoid these issues, the specification should aim to contain a written description of the invention in a way that it is understandable, full, clear and concise126,127.
Challenges in ethical and reliable AI
Transparency and explainability are crucial for regulatory approval and clinical adoption124. Lack of transparency and interpretability in AI models, particularly black-box systems, complicates the adoption and fostering of trust in employing AI in drug development128,129. Explainability refers to the AI’s ability to clarify its decision-making process and bias mitigation methods in a manner that is comprehensible to humans120. This transparency facilitates the determination of liability in cases of adverse outcomes. This transparency is especially vital in clinical trials, where regulatory bodies must understand AI-driven predictions to assess their validity and ensure patient safety. Without clear explanations, AI-generated drug candidates may face increased scrutiny, delaying trial approvals and regulatory acceptance130. While regulations remain underdeveloped, guidelines such as DECIDE-AI, SPIRIT-AI and CONSORT-AI provide structured reporting frameworks to enhance accountability in AI-driven clinical research131–133. However, these guidelines primarily focus on AI applications in disease diagnosis rather than drug development. The increasing integration of AI into drug development underscores the urgent need for algorithmic inclusivity and transparency. Policymakers must attempt to translate these guidelines to standardized regulations to protect patients from associated risks.
The quality and efficiency of AI-led drug discovery are heavily contingent on data reliability, representativeness and diversity. Limited or inconsistent training data can impair model performance by misrepresenting the molecular structure of drugs134. Even minor alterations in chemical structure can significantly impact binding affinity, pharmacokinetics and pharmacodynamics, potentially rendering therapies ineffective or toxic134. Several solutions may be considered to overcome these challenges. Firstly, a centralized biomarker repository with standardized and nationally representative data may minimize inconsistencies and improve representation124. Secondly, transfer learning, which applies knowledge from related tasks, can improve learning efficiency with limited datasets. Thirdly, GenAI models, such as generative adversarial networks (GANs) and variational autoencoder (VAEs), can generate synthetic data to overcome data scarcity. For example, MedGAN, a DL model combining GANs and graph convolutional networks facilitates the generation of new quinoline-scaffold molecules135. This approach may be particularly attractive for rare ophthalmic diseases, where real-world data are limited and synthetic augmentation could help improve dataset diversity. However, GANs also bring their own set of challenges, including the risk of generating biologically irrelevant or harmful compounds, limitations in transparency and subsequent regulatory acceptance, amplified bias, and training instability. Without rigorous validation, synthetic data may inadvertently reinforce existing biases or produce misleading outputs—especially in disease areas with incomplete biological understanding. These risks highlight the need for transparency, oversight, and cautious interpretation in applying synthetic data to rare disease research. Without a standardized approach to integrating AI-generated candidates into established drug pipelines, regulatory acceptance will remain a significant barrier.
Ensuring equitable and ethical use of AI in drug development is another key consideration. AI models can inadvertently perpetuate bias and discrimination124. For instance, VisionFM, a foundation model that fosters multiple AI applications including ophthalmological diagnosis and biomarker identification, was pretrained on a dataset disproportionately representing the Chinese population88. Consequently, it may fail to capture the heterogeneity of rare conditions such as Leber hereditary optic neuropathy, Best disease and Bietti Crystalline Dystrophy136. If such biases extend to AI-driven drug discovery, they could result in therapies that are less effective for underrepresented populations, ultimately skewing clinical trial results and limiting the generalizability of findings137. Moreover, access to cutting-edge AI tools and computational infrastructure remains uneven across regions, impacting low- and middle-income countries (LMICs) the most, hindering their ability to participate in AI-driven research. This exacerbates healthcare inequity where AI-generated treatments may fail to adequately address the health needs of these populations. LMICs often face systemic barriers—such as fragmented health systems, limited biobank access, and underrepresentation in genomic datasets—which further restrict their participation in AI-driven biomedical research and innovation138. Bias in training data can also impact participant selection in AI-optimized trial recruitment, leading to skewed demographic representation and potentially confounded results. Current AI-assisted recruitment strategies have shown promise in streamlining patient matching based on eligibility criteria, yet they often rely on EHRs that may underrepresent certain populations. Similarly, digital twins must be constructed using high-quality, representative datasets. If built on biased or incomplete data, digital twins may yield misleading predictions, potentially leading to erroneous trial designs or ineffective drug candidates. Data augmentation offers a potential solution by generating synthetic data and enhancing data diversity, thereby mitigating discrimination in AI-powered drug discovery. However, data augmentation alone cannot fully address biases; robust validation protocols and continuous monitoring are necessary to ensure fairness and accuracy in clinical trials137.
As AI increasingly shapes drug discovery, regulatory updates must address these emerging applications139. Given AI’s transformative impact, industry stakeholders must enhance quality assessment beyond FDA oversight123. Ensuring equity in AI-driven drug development, establishing patent frameworks, and promoting transparency will be critical to widespread adoption.
Conclusions
AI has a huge potential to transform drug discovery and development, offering groundbreaking opportunities in molecular drug target identification, preclinical and clinical testing, and drug repurposing. However, the ethical and societal complexities inherent in this technology necessitate careful and deliberate strategies to fully leverage its benefits120. As AI continues to evolve, it is essential to stay informed about emerging developments and implement responsible practices to ensure equitable benefits for all patients120.
Acknowledgements
J.L.G. and V.B.M. gratefully acknowledge support from the National Eye Institute (P30-EY026877) and Research to Prevent Blindness, Inc. The funders had no roles in in the conception, execution or reporting of this review. T.Y.W. is supported by National Key R&D Program (Grant No.: 2022YFC2502800), National Natural Science Fund of China (Grant No.: 82388101) and Beijing Natural Science Foundation (Grant No.: IS23096). D.S.W.T. is supported by the National Medical Research Council, Singapore (MOH-001689-00, MOH-000655-00 and MOH-001014-00), the Duke-NUS Medical School (Duke-NUS/RSF/2021/0018 and 05/FY2020/EX/15-A58, 05/FY2022/EX/66-A128) and the Agency for Science, Technology and Research (A20H4g2141 and H20C6a0032). D.S.J.T. acknowledges support from the Medical Research Council (MRC) Clinician Scientist Fellowship (UKRI2441), the MRC Proximity to Discovery and Impact (P2D) Fund (MR/X502996/1), and the Birmingham Health Partners Clinician Scientist Fellowship.
Author contributions
H.C., D.S.J.T., and D.S.W.T. conceptualized the paper. H.C., J.W., and C.Q. wrote the main manuscript text and prepared Figs. 1–4 and Tables 1–3. J.G., V.M., T.Y.W., J.M., D.S.W.T., and D.S.J.T. edited all revisions of the manuscript. All authors approved the final manuscript.
Data Availability
No datasets were generated or analysed during the current study.
Competing interests
D.S.W.T. holds a patent on a deep learning system for the detection of retinal diseases (10201706186V) and a computer-implemented method for training an image classifier using weakly annotated training data (10201901083Y) and stocks at EyRIS, Singapore and aISIGHT. The other authors declare no conflicts of interest.
Footnotes
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Contributor Information
Daniel S. W. Ting, Email: daniel.ting45@gmail.com
Darren S. J. Ting, Email: d.s.j.ting@bham.ac.uk
References
- 1.Sertkaya, A., Beleche, T., Jessup, A. & Sommers, B. D. Costs of drug development and research and development intensity in the US, 2000-2018. JAMA Netw. Open7, e2415445 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Spherical Insights LLP. Global Ophthalmic Drugs Market Size, Share, and COVID-19 Impact Analysis, by Disease, Drug Class, Dosage, Administration, Product, and Region, Forecast to 2030. (published February 2023; accessed 20 August 2025).
- 3.Scott, I. U. et al. A phase II randomized clinical trial of intravitreal bevacizumab for diabetic macular edema. Ophthalmology114, 1860–1867 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Rosenfeld, P. J., Moshfeghi, A. A. & Puliafito, C. A. Optical coherence tomography findings after an intravitreal injection of bevacizumab (avastin) for neovascular age-related macular degeneration. Ophthalmic Surg. Lasers Imaging36, 331–335 (2005). [PubMed] [Google Scholar]
- 5.Cheung, N., Mitchell, P. & Wong, T. Y. Diabetic retinopathy. Lancet376, 124–136 (2010). [DOI] [PubMed] [Google Scholar]
- 6.Wong, T. Y., Cheung, C. M. G., Larsen, M., Sharma, S. & Simó, R. Diabetic retinopathy. Nat. Rev. Dis. Prim.2, 16012 (2016). [DOI] [PubMed] [Google Scholar]
- 7.Mitchell, P., Liew, G., Gopinath, B. & Wong, T. Y. Age-related macular degeneration. Lancet392, 1147–1159 (2018). [DOI] [PubMed] [Google Scholar]
- 8.Bloch, S. B., Larsen, M. & Munch, I. C. Incidence of legal blindness from age-related macular degeneration in Denmark: year 2000 to 2010. Am. J. Ophthalmol.153, 209–213.e202 (2012). [DOI] [PubMed] [Google Scholar]
- 9.Israeli, A., Peer, H., Gleitman, I., Falick, Y. & Mezer, E. Time trends of etiologies of blindness in Israel 2009-2020: have methods to decrease leading causes of blindness been exhausted?. Am. J. Ophthalmol.240, 149–158 (2022). [DOI] [PubMed] [Google Scholar]
- 10.Russell, S. et al. Efficacy and safety of voretigene neparvovec (AAV2-hRPE65v2) in patients with RPE65-mediated inherited retinal dystrophy: a randomised, controlled, open-label, phase 3 trial. Lancet390, 849–860 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Bourne, R. R. A. et al. The UK clinical eye research strategy: refreshing research priorities for clinical eye research in the UK. Eye38, 1947–1957 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Burki, T. A new paradigm for drug development. Lancet Digit. Health2, e226–e227 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Stokes, J. M. et al. A deep learning approach to antibiotic discovery. Cell180, 688–702.e613 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Ho, C. S. et al. Antimicrobial resistance: a concise update. Lancet Microbe6, 100947. 10.1016/j.lanmic.2024.07.010 (2025). [DOI] [PubMed] [Google Scholar]
- 15.Mock, M., Edavettal, S., Langmead, C. & Russell, A. AI can help to speed up drug discovery—but only if we give it the right data. Nature621, 467–470 (2023). [DOI] [PubMed] [Google Scholar]
- 16.Liu, Z. et al. AI-based language models powering drug discovery and development. Drug Discov. Today26, 2593–2607 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Jeyaraman, M. et al. ChatGPT in action: harnessing artificial intelligence potential and addressing ethical challenges in medicine, education, and scientific research. World J. Methodol.13, 170–178 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Thirunavukarasu, A. J. et al. Large language models in medicine. Nat. Med.29, 1930–1940 (2023). [DOI] [PubMed] [Google Scholar]
- 19.Zhang, K. et al. Artificial intelligence in drug development. Nat. Med.31, 45–59 (2025). [DOI] [PubMed] [Google Scholar]
- 20.Harrer, S., Shah, P., Antony, B. & Hu, J. Artificial intelligence for clinical trial design. Trends Pharmacol. Sci.40, 577–591 (2019). [DOI] [PubMed] [Google Scholar]
- 21.Liao, Q. et al. Application of artificial intelligence in drug-target interactions prediction: a review. npj Biomed. Innov.2, 1 (2025). [Google Scholar]
- 22.Sadybekov, A. V. & Katritch, V. Computational approaches streamlining drug discovery. Nature616, 673–685 (2023). [DOI] [PubMed] [Google Scholar]
- 23.Weinberg, J., Gaur, M., Swaroop, A. & Taylor, A. Proteostasis in aging-associated ocular disease. Mol. Asp. Med.88, 101157 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Venkatraman, A., Duong-Thi, M. D., Pervushin, K., Ohlson, S. & Mehta, J. S. Pharmaceutical modulation of the proteolytic profile of Transforming Growth Factor Beta induced protein (TGFBIp) offers a new avenue for treatment of TGFBI-corneal dystrophy. J. Adv. Res.24, 529–543 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Burley, S. K. et al. Protein Data Bank: a comprehensive review of 3D structure holdings and worldwide utilization by researchers, educators, and students. Biomolecules12, 10.3390/biom12101425 (2022). [DOI] [PMC free article] [PubMed]
- 26.Senior, A. W. et al. Improved protein structure prediction using potentials from deep learning. Nature577, 706–710 (2020). [DOI] [PubMed] [Google Scholar]
- 27.Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. Nature596, 583–589 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Abramson, J. et al. Accurate structure prediction of biomolecular interactions with AlphaFold 3. Nature630, 493–500 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Zhang, Z. et al. Structure prediction of novel isoforms from uveal melanoma by AlphaFold. Sci. Data10, 513 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Kamya, P. et al. PandaOmics: an AI-driven platform for therapeutic target and biomarker discovery. J. Chem. Inf. Model.64, 3961–3969 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Wan, F., Torres, M. D. T., Peng, J. & de la Fuente-Nunez, C. Deep-learning-enabled antibiotic discovery through molecular de-extinction. Nat. Biomed. Eng.8, 854–871 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Maasch, J., Torres, M. D. T., Melo, M. C. R. & de la Fuente-Nunez, C. Molecular de-extinction of ancient antimicrobial peptides enabled by machine learning. Cell Host Microbe31, 1260–1274.e1266 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Torres, M. D. T. et al. Mining human microbiomes reveals an untapped source of peptide antibiotics. Cell187, 5453–5467.e5415 (2024). [DOI] [PubMed] [Google Scholar]
- 34.Santos-Júnior, C. D. et al. Discovery of antimicrobial peptides in the global microbiome with machine learning. Cell187, 3761–3778.e3716 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Redshaw, J., Ting, D. S. J., Brown, A., Hirst, J. D. & Gärtner, T. Krein support vector machine classification of antimicrobial peptides. Digital Discov.2, 502–511 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Gentile, F. et al. Deep docking: a deep learning platform for augmentation of structure based drug discovery. ACS Cent. Sci.6, 939–949 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Luttens, A. et al. Rapid traversal of vast chemical space using machine learning-guided docking screens. Nat. Comput. Sci. 10.1038/s43588-025-00777-x (2025). [DOI] [PMC free article] [PubMed]
- 38.Ren, F. et al. A small-molecule TNIK inhibitor targets fibrosis in preclinical and clinical models. Nat. Biotechnol.10.1038/s41587-024-02143-0 (2024). [DOI] [PMC free article] [PubMed]
- 39.Hollingsworth, S. A. & Dror, R. O. Molecular dynamics simulation for all. Neuron99, 1129–1143 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Prašnikar, E., Ljubič, M., Perdih, A. & Borišek, J. Machine learning heralding a new development phase in molecular dynamics simulations. Artif. Intell. Rev.57, 102 (2024). [Google Scholar]
- 41.Ting, D. S. J. et al. Evaluation of host defense peptide (CaD23)-antibiotic interaction and mechanism of action: insights from experimental and molecular dynamics simulations studies. Front. Pharmacol.12, 10.3389/fphar.2021.731499 (2021). [DOI] [PMC free article] [PubMed]
- 42.Ting, D. S. J., Lakshminarayanan, R., Mohammad, I. & Dua, H. Development of novel human-derived hybrid host defense peptides for infectious keratitis. BMJ Open Ophthalmol.8, (2023).
- 43.Tropsha, A., Isayev, O., Varnek, A., Schneider, G. & Cherkasov, A. Integrating QSAR modelling and deep learning in drug discovery: the emergence of deep QSAR. Nat. Rev. Drug Discov.23, 141–155 (2024). [DOI] [PubMed] [Google Scholar]
- 44.Swanson, K. et al. Generative AI for designing and validating easily synthesizable and structurally novel antibiotics. Nat. Mach. Intell.6, 338–353 (2024). [Google Scholar]
- 45.Das, P. et al. Accelerated antimicrobial discovery via deep generative models and molecular dynamics simulations. Nat. Biomed. Eng.5, 613–623 (2021). [DOI] [PubMed] [Google Scholar]
- 46.Turon, G. et al. First fully-automated AI/ML virtual screening cascade implemented at a drug discovery centre in Africa. Nat. Commun.14, 5736 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Alizadehsani, R. et al. Explainable artificial intelligence for drug discovery and development: a comprehensive survey. IEEE Access12, 35796–35812 (2024). [Google Scholar]
- 48.Jiménez-Luna, J., Grisoni, F. & Schneider, G. Drug discovery with explainable artificial intelligence. Nat. Mach. Intell.2, 573–584 (2020). [Google Scholar]
- 49.Madushanka, A., Laird, E., Clark, C. & Kraka, E. SmartCADD: AI-QM empowered drug discovery platform with explainability. J. Chem. Inf. Model.64, 6799–6813 (2024). [DOI] [PubMed] [Google Scholar]
- 50.OpenAI et al. OpenAI o1 System Card. arXivhttps://ui.adsabs.harvard.edu/abs/2024arXiv241216720O (2024).
- 51.Team, G. et al. Gemini: a family of highly capable multimodal models. arXivhttps://ui.adsabs.harvard.edu/abs/2023arXiv231211805G (2023).
- 52.DeepSeek-AI et al. DeepSeek-R1: incentivizing reasoning capability in LLMs via reinforcement learning. arXivhttps://ui.adsabs.harvard.edu/abs/2025arXiv250112948D (2025).
- 53.Loewa, A., Feng, J. J. & Hedtrich, S. Human disease models in drug development. Nat. Rev. Bioeng.1, 545–559 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Shi, H. et al. Organoid intelligence: integration of organoid technology and artificial intelligence in the new era of in vitro models. Med. Nov. Technol. Devices21, 100276 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Achberger, K. et al. Merging organoid and organ-on-a-chip technology to generate complex multi-layer tissue models in a human retina-on-a-chip platform. eLife8, e46188 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Chung, M. et al. Wet-AMD on a chip: modeling outer blood-retinal barrier in vitro. Adv. Healthc. Mater.7, 1700028 (2018). [DOI] [PubMed] [Google Scholar]
- 57.Seo, J. et al. Multiscale reverse engineering of the human ocular surface. Nat. Med.25, 1310–1318 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Choi, D.-H. et al. Analyzing angiogenesis on a chip using deep learning-based image processing. Lab Chip23, 475–484 (2023). [DOI] [PubMed] [Google Scholar]
- 59.Dressler, O. J., Howes, P. D., Choo, J. & deMello, A. J. Reinforcement learning for dynamic microfluidic control. ACS Omega3, 10084–10091 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Schuster, B. et al. Automated microfluidic platform for dynamic and combinatorial drug screening of tumor organoids. Nat. Commun.11, 5271 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Tebon, P. J. et al. Drug screening at single-organoid resolution via bioprinting and interferometry. Nat. Commun.14, 3168 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Mukashyaka, P. et al. High-throughput deconvolution of 3D organoid dynamics at cellular resolution for cancer pharmacology with Cellos. Nat. Commun.14, 8406 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kuepfer, L. et al. Applied concepts in PBPK modeling: how to build a PBPK/PD model. CPT Pharmacomet. Syst. Pharm.5, 516–531 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Jones, H. & Rowland-Yeo, K. Basic concepts in physiologically based pharmacokinetic modeling in drug discovery and development. CPT Pharmacomet. Syst. Pharm.2, e63 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Wang, N. et al. How can machine learning and multiscale modeling benefit ocular drug development?. Adv. Drug Deliv. Rev.196, 114772 (2023). [DOI] [PubMed] [Google Scholar]
- 66.Kompella, U. B., Hartman, R. R. & Patil, M. A. Extraocular, periocular, and intraocular routes for sustained drug delivery for glaucoma. Prog. Retinal Eye Res.82, 100901 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Ting, D. S. J. et al. Hybrid derivative of cathelicidin and human beta defensin-2 against Gram-positive bacteria: a novel approach for the treatment of bacterial keratitis. Sci. Rep.11, 18304 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Chou, W.-C. & Lin, Z. Machine learning and artificial intelligence in physiologically based pharmacokinetic modeling. Toxicol. Sci.191, 1–14 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Lu, J., Deng, K., Zhang, X., Liu, G. & Guan, Y. Neural-ODE for pharmacokinetics modeling and its advantage to alternative machine learning models in predicting new dosing regimens. iScience24, 102804 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Liu, X. et al. Long short-term memory recurrent neural network for pharmacokinetic-pharmacodynamic modeling. Int. J. Clin. Pharm. Ther.59, 138–146 (2021). [DOI] [PubMed] [Google Scholar]
- 71.Szustakowski, J. D. et al. Advancing human genetics research and drug discovery through exome sequencing of the UK Biobank. Nat. Genet.53, 942–948 (2021). [DOI] [PubMed] [Google Scholar]
- 72.Zhou, W., Li, X., Han, L. & Fan, S. in Network Pharmacology (ed. Li, S.) 35–73 (Springer, 2021).
- 73.Allesøe, R. L. et al. Discovery of drug–omics associations in type 2 diabetes with generative deep-learning models. Nat. Biotechnol.41, 399–408 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Wolf, J. et al. Using electronic health record data to determine the safety of aqueous humor liquid biopsies for molecular analyses. Ophthalmol. Sci.4, 100517 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Wolf, J. et al. Liquid biopsy proteomics in ophthalmology. J. Proteome Res.23, 511–522 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Kato, Y. et al. Age-related maculopathy susceptibility 2 and complement factor H polymorphism and intraocular complement activation in neovascular age-related macular degeneration. Ophthalmol. Sci.2, 10.1016/j.xops.2022.100167 (2022). [DOI] [PMC free article] [PubMed]
- 77.Wolf, J. et al. The Human Eye Transcriptome Atlas: a searchable comparative transcriptome database for healthy and diseased human eye tissue. Genomics114, 110286 (2022). [DOI] [PubMed] [Google Scholar]
- 78.Alli-Shaik, A. et al. System-wide vitreous proteome dissection reveals impaired sheddase activity in diabetic retinopathy. Theranostics12, 6682–6704 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Wolf, J. et al. Liquid-biopsy proteomics combined with AI identifies cellular drivers of eye aging and disease in vivo. Cell186, 4868–4884.e4812 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Hallam, T. M. et al. Ocular biomarker profiling after complement factor I gene therapy in geographic atrophy secondary to age-related macular degeneration. medRxiv10.1101/2024.06.12.24308796 (2024).
- 81.He, D., Liu, Q., Wu, Y. & Xie, L. A context-aware deconfounding autoencoder for robust prediction of personalized clinical drug response from cell-line compound screening. Nat. Mach. Intell.4, 879–892 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Beykin, G., Norcia, A. M., Srinivasan, V. J., Dubra, A. & Goldberg, J. L. Discovery and clinical translation of novel glaucoma biomarkers. Prog. Retin. Eye Res.80, 100875 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Beykin, G. & Goldberg, J. L. Molecular biomarkers for glaucoma. Curr. Ophthalmol. Rep.7, 171–176 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Norcia, A. M., Yakovleva, A., Jehangir, N. & Goldberg, J. L. Preferential loss of contrast decrement responses in human glaucoma. Invest. Ophthalmol. Vis. Sci.63, 16 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Zhou, Y. et al. AutoMorph: automated retinal vascular morphology quantification via a deep learning pipeline. Transl. Vis. Sci. Technol.11, 12–12 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Moraes, G. et al. Quantitative analysis of OCT for neovascular age-related macular degeneration using deep learning. Ophthalmology128, 693–705 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Li, J. O. et al. Digital technology, tele-medicine and artificial intelligence in ophthalmology: a global perspective. Prog. Retin. Eye Res.82, 100900 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Qiu, J. et al. Development and Validation of a Multimodal Multitask Vision Foundation Model for Generalist Ophthalmic Artificial Intelligence. NEJM AI1, AIoa2300221, 10.1056/AIoa2300221 (2024). [Google Scholar]
- 89.Xiong, F. et al. Ocular signs and testing most compatible with sarcoidosis-associated uveitis: a latent class analysis. Ophthalmol. Sci.4, 10.1016/j.xops.2024.100503 (2024). [DOI] [PMC free article] [PubMed]
- 90.Holland, R. et al. Deep-learning-based clustering of OCT images for biomarker discovery in age-related macular degeneration (PINNACLE study report 4). Ophthalmol. Sci.10.1016/j.xops.2024.100543 (2024). [DOI] [PMC free article] [PubMed]
- 91.Moraes, G. et al. Quantifying changes on OCT in eyes receiving treatment for neovascular age-related macular degeneration. Ophthalmol. Sci.4, 10.1016/j.xops.2024.100570 (2024). [DOI] [PMC free article] [PubMed]
- 92.Massengill, M. T., Cubillos, S., Sheth, N., Sethi, A. & Lim, J. I. Response of diabetic macular edema to anti-VEGF medications correlates with improvement in macular vessel architecture measured with OCT angiography. Ophthalmol. Sci.4, 10.1016/j.xops.2024.100478 (2024). [DOI] [PMC free article] [PubMed]
- 93.Gallardo, M. et al. Machine learning can predict anti–VEGF treatment demand in a treat-and-extend regimen for patients with neovascular AMD, DME, and RVO associated macular edema. Ophthalmol. Retin.5, 604–624 (2021). [DOI] [PubMed] [Google Scholar]
- 94.Zhao, X. et al. Optical coherence tomography-based short-term effect prediction of anti-vascular endothelial growth factor treatment in neovascular age-related macular degeneration using sensitive structure guided network. Graefes Arch. Clin. Exp. Ophthalmol.259, 3261–3269 (2021). [DOI] [PubMed] [Google Scholar]
- 95.Lee, H., Kim, S., Kim, M. A., Chung, H. & Kim, H. C. Post-treatment prediction of optical coherence tomography using a conditional generative adversarial network in age-related macular degeneration. Retina41, 572–580 (2021). [DOI] [PubMed] [Google Scholar]
- 96.Schmetterer, L. et al. Endpoints for clinical trials in ophthalmology. Prog. Retin. Eye Res.97, 101160 (2023). [DOI] [PubMed] [Google Scholar]
- 97.Sun, M. T. et al. Structural and metabolic imaging after short-term use of the balance goggles system in glaucoma patients: a pilot study. J. Glaucoma31, 634–638 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Levin, L. A., Patrick, C., Choudry, N. B., Sharif, N. A. & Goldberg, J. L. Neuroprotection in neurodegenerations of the brain and eye: lessons from the past and directions for the future. Front. Neurol.13, 964197 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Wareham, L. K. et al. Solving neurodegeneration: common mechanisms and strategies for new treatments. Mol. Neurodegener.17, 23 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Williamson, D. J. et al. Artificial intelligence to facilitate clinical trial recruitment in age-related macular degeneration. Ophthalmol. Sci.4, 100566 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Vayena, E., Blasimme, A. & Sugarman, J. Decentralised clinical trials: ethical opportunities and challenges. Lancet Digital Health5, e390–e394 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Teo, Z. L. et al. Federated machine learning in healthcare: a systematic review on clinical applications and technical architecture. Cell Rep. Med.5, 10.1016/j.xcrm.2024.101419 (2024). [DOI] [PMC free article] [PubMed]
- 103.Pallmann, P. et al. Adaptive designs in clinical trials: why use them, and how to run and report them. BMC Med.16, 29 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Huckvale, K. et al. Protocol for a bandit-based response adaptive trial to evaluate the effectiveness of brief self-guided digital interventions for reducing psychological distress in university students: the Vibe Up study. BMJ Open13, e066249 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Vidovszky, A. A. et al. Increasing acceptance of AI-generated digital twins through clinical trial applications. Clin. Transl. Sci.17, e13897 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Bordukova, M., Makarov, N., Rodriguez-Esteban, R., Schmich, F. & Menden, M. P. Generative artificial intelligence empowers digital twins in drug discovery and clinical trials. Expert Opin. Drug Discov.19, 33–42 (2024). [DOI] [PubMed] [Google Scholar]
- 107.Visioli, G. et al. Proliferative vitreoretinopathy in retinal detachment: perspectives on building a digital twin model using nintedanib. Int. J. Mol. Sci.25, 11074 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Iliuţă, M.-E. et al. Digital twin models for personalised and predictive medicine in ophthalmology. Technologies12, 55 (2024). [Google Scholar]
- 109.Li, F. et al. Development and clinical deployment of a smartphone-based visual field deep learning system for glaucoma detection. npj Digital Med.3, 123 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Chen, W. et al. Early detection of visual impairment in young children using a smartphone-based deep learning system. Nat. Med.29, 493–503 (2023). [DOI] [PubMed] [Google Scholar]
- 111.Li, J. et al. Integrated image-based deep learning and language models for primary diabetes care. Nat. Med.10.1038/s41591-024-03139-8 (2024). [DOI] [PMC free article] [PubMed]
- 112.Zong, N. et al. Computational drug repurposing based on electronic health records: a scoping review. npj Digital Med.5, 77 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Liu, R., Wei, L. & Zhang, P. A deep learning framework for drug repurposing via emulating clinical trials on real-world patient data. Nat. Mach. Intell.3, 68–75 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Beck, B. R., Shin, B., Choi, Y., Park, S. & Kang, K. Predicting commercially available antiviral drugs that may act on the novel coronavirus (SARS-CoV-2) through a drug-target interaction deep learning model. Comput. Struct. Biotechnol. J.18, 784–790 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Zhou, Y., Wang, F., Tang, J., Nussinov, R. & Cheng, F. Artificial intelligence in COVID-19 drug repurposing. Lancet Digital Health2, e667–e676 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Ting, D. S. J., Tan, T. F. & Ting, D. S. W. ChatGPT in ophthalmology: the dawn of a new era?. Eye38, 4–7 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Thirunavukarasu, A. J. et al. Large language models approach expert-level clinical knowledge and reasoning in ophthalmology: a head-to-head cross-sectional study. PLoS Digit. Health3, e0000341 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Yan, C. et al. Leveraging generative AI to prioritize drug repurposing candidates for Alzheimer’s disease with real-world clinical validation. npj Digital Med.7, 46 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Ouyang, B. et al. AI-powered omics-based drug pair discovery for pyroptosis therapy targeting triple-negative breast cancer. Nat. Commun.15, 7560 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Katwaroo, A. R., Adesh, V. S., Lowtan, A. & Umakanthan, S. The diagnostic, therapeutic, and ethical impact of artificial intelligence in modern medicine. Postgrad. Med. J.100, 289–296 (2023). [DOI] [PubMed] [Google Scholar]
- 121.Narmadha, K. & Varalakshmi, P. Federated Learning in Healthcare: a privacy preserving approach. Stud. Health Technol. Inf.294, 194–198 (2022). [DOI] [PubMed] [Google Scholar]
- 122.U.S. Food & Drug Administration. Artificial Intelligence in Software as a Medical Device (SaMD). FDA (published 25 March 2025; accessed 20 August 2025).
- 123.Warraich, H. J., Tazbaz, T. & Califf, R. M. FDA perspective on the regulation of artificial intelligence in health care and biomedicine. JAMA333, 241–247 (2025). [DOI] [PubMed] [Google Scholar]
- 124.Mirakhori, F. & Niazi, S. K. Harnessing the AI/ML in drug and biological products discovery and development: the regulatory perspective. Pharmaceuticals18, 47 (2025). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Hutson, M. AI for drug discovery is booming, but who owns the patents?. Nat. Biotechnol.41, 1494–1496 (2023). [DOI] [PubMed] [Google Scholar]
- 126.Krumplitsch, S. Generative AI, drug discovery, and US patent law. https://www.dlapiper.com/en-cl/insights/publications/synthesis/2024/generative-ai-drug-discovery-and-us-patent-law (2024).
- 127.School, C. L. 35 U.S. Code § 112—Specification. https://www.law.cornell.edu/uscode/text/35/112 (2024).
- 128.Keskinbora, K. H. Medical ethics considerations on artificial intelligence. J. Clin. Neurosci.64, 277–282 (2019). [DOI] [PubMed] [Google Scholar]
- 129.Ong, Z. Z. et al. Diagnostic performance of deep learning for infectious keratitis: a systematic review and meta-analysis. EClinicalMedicine77, 102887 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Inglada Galiana, L., Corral Gudino, L. & Miramontes González, P. Ethics and artificial intelligence. Rev. Clín. Esp.224, 178–186 (2024). [DOI] [PubMed] [Google Scholar]
- 131.Vasey, B. et al. Reporting guideline for the early-stage clinical evaluation of decision support systems driven by artificial intelligence: DECIDE-AI. Nat. Med.28, 924–933 (2022). [DOI] [PubMed] [Google Scholar]
- 132.Liu, X. et al. Reporting guidelines for clinical trial reports for interventions involving artificial intelligence: the CONSORT-AI extension. Nat. Med.26, 1364–1374 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Cruz Rivera, S. et al. Guidelines for clinical trial protocols for interventions involving artificial intelligence: the SPIRIT-AI extension. Nat. Med.26, 1351–1363 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Gangwal, A., Ansari, A., Ahmad, I., Azad, A. K. & Wan Sulaiman, W. M. A. Current strategies to address data scarcity in artificial intelligence-based drug discovery: a comprehensive review. Comput. Biol. Med.179, 108734 (2024). [DOI] [PubMed] [Google Scholar]
- 135.Macedo, B., Ribeiro Vaz, I. & Taveira Gomes, T. MedGAN: optimized generative adversarial network with graph convolutional networks for novel molecule design. Sci. Rep.14, 1212 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Tiwari, P. C., Pal, R., Chaudhary, M. J. & Nath, R. Artificial intelligence revolutionizing drug development: exploring opportunities and challenges. Drug Dev. Res.84, 1652–1663 (2023). [DOI] [PubMed] [Google Scholar]
- 137.Blanco-González, A. et al. The role of AI in drug discovery: challenges, opportunities, and strategies. Pharmaceuticals16, 891 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Jalali, R., Nogueira-Rodrigues, A., Das, A., Sirohi, B. & Panda, P. K. Drug development in low- and middle-income countries: opportunity or exploitation? Am. Soc. Clin. Oncol. Educ. Book 3–10 (2022). [DOI] [PubMed]
- 139.Ibrahim, H. et al. Reporting guidelines for clinical trials of artificial intelligence interventions: the SPIRIT-AI and CONSORT-AI guidelines. Trials22, 11 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Sands, B. E. et al. Phase 2 trial of anti-TL1A monoclonal antibody tulisokibart for ulcerative colitis. N. Engl. J. Med.391, 1119–1129 (2024). [DOI] [PubMed] [Google Scholar]
- 141.Li, Y. et al. Abstract 502: ISM3091, a novel selective USP1 inhibitor as a targeted anticancer therapy. Cancer Res.83, 502–502 (2023). [Google Scholar]
- 142.Insilico. A Small Molecule KAT6 Inhibitor as a Potential Treatment of ER+ HER2- Breast Cancer (Entering IND). https://insilico.com/pipeline_target_kat6 (2024).
- 143.Ren, F. et al. A small-molecule TNIK inhibitor targets fibrosis in preclinical and clinical models. Nat. Biotechnol.43, 63–75 (2025). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Booq, R. Y., Tawfik, E. A., Alfassam, H. A., Alfahad, A. J. & Alyamani, E. J. Assessment of the antibacterial efficacy of halicin against pathogenic bacteria. Antibiotics10, 10.3390/antibiotics10121480 (2021). [DOI] [PMC free article] [PubMed]
- 145.Treat, J. et al. P2.12-07 phase II trial of LP-300 + pemetrexed/carboplatin in patients who never smoked with lung adenocarcinoma after TKI progression. J. Thorac. Oncol.18, S365 (2023). [Google Scholar]
- 146.Kulkarni, A. et al. LP-184, a novel acylfulvene molecule, exhibits anticancer activity against diverse solid tumors with homologous recombination deficiency. Cancer Res. Commun.4, 1199–1210 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Zhou, J. et al. LP-284, a small molecule acylfulvene, exerts potent antitumor activity in preclinical non-Hodgkin’s lymphoma models and in cells deficient in DNA damage repair. Oncotarget14, 597–611 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Schmidt-Erfurth, U. et al. Therapeutic response in the HAWK and HARRIER trials using deep learning in retinal fluid volume and compartment analysis. Eye37, 1160–1169 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Fu, D. J. et al. Deep-learning automated quantification of longitudinal OCT scans demonstrates reduced RPE loss rate, preservation of intact macular area and predictive value of isolated photoreceptor degeneration in geographic atrophy patients receiving C3 inhibition treatment. Br. J. Ophthalmol.108, 536–545 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Vogl, W.-D. et al. Predicting topographic disease progression and treatment response of pegcetacoplan in geographic atrophy quantified by deep learning. Ophthalmol. Retin.7, 4–13 (2023). [DOI] [PubMed] [Google Scholar]
- 151.Roberts, P. K. et al. Quantification of fluid resolution and visual acuity gain in patients with diabetic macular edema using deep learning: a post hoc analysis of a randomized clinical trial. JAMA Ophthalmol.138, 945–953 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Schmidt-Erfurth, U. et al. Disease activity and therapeutic response to pegcetacoplan for geographic atrophy identified by deep learning-based analysis of OCT. Ophthalmology10.1016/j.ophtha.2024.08.017 (2024). [DOI] [PubMed]
- 153.Mulyukov, Z. et al. Artificial intelligence-based disease activity monitoring to personalized neovascular age-related macular degeneration treatment: a feasibility study. Ophthalmol. Sci.4, 100565 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Chandra, R. S. & Ying, G. S. Predicting visual acuity responses to anti-VEGF treatment in the comparison of age-related macular degeneration treatments trials using machine learning. Ophthalmol. Retin.8, 419–430 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Kikuchi, Y. et al. Machine learning to predict faricimab treatment outcome in neovascular age-related macular degeneration. Ophthalmol. Sci.4, 10.1016/j.xops.2023.100385 (2024). [DOI] [PMC free article] [PubMed]
- 156.Maunz, A. et al. Machine learning to predict response to ranibizumab in neovascular age-related macular degeneration. Ophthalmol. Sci.3, 100319 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Bogunović, H. et al. Prediction of anti-VEGF treatment requirements in neovascular AMD using a machine learning approach. Invest. Ophthalmol. Vis. Sci.58, 3240–3248 (2017). [DOI] [PubMed] [Google Scholar]
- 158.Chandra, R. S. & Ying, G. S. Evaluation of multiple machine learning models for predicting number of anti-VEGF injections in the comparison of AMD treatment trials (CATT). Transl. Vis. Sci. Technol.12, 18 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No datasets were generated or analysed during the current study.




