Next Article in Journal
Quercetin Nanocrystal Gel: A Novel Topical Therapeutic Strategy for Androgenetic Alopecia
Previous Article in Journal
Development of pH-Sensitive Multiparticulates for Orally Disintegrating Tablets of Proton Pump Inhibitors: Physicochemical Characterization and Drug Release Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Machine Learning for Multi-Target Drug Discovery: Challenges and Opportunities in Systems Pharmacology

1
Department of Pharmacy, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
2
School of Physics and Electronic Information, Yan’an University, Yan’an 716000, China
3
Yan’an Medical College of Yan’an University, Yan’an 716000, China
4
Department of Science and Education, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
*
Author to whom correspondence should be addressed.
Pharmaceutics 2025, 17(9), 1186; https://doi.org/10.3390/pharmaceutics17091186
Submission received: 18 August 2025 / Revised: 5 September 2025 / Accepted: 11 September 2025 / Published: 12 September 2025
(This article belongs to the Special Issue Advanced Algorithms for Small-Molecule Therapeutics Development)

Abstract

Multi-target drug discovery has become an essential strategy for treating complex diseases involving multiple molecular pathways. Traditional single-target approaches often fall short in addressing the multifactorial nature of conditions such as cancer and neurodegenerative disorders. With the rise in large-scale biological data and algorithmic advances, machine learning (ML) has emerged as a powerful tool to accelerate and optimize multi-target drug development. This review presents a comprehensive overview of ML techniques, including advanced deep learning (DL) approaches like attention-based models, and highlights their application in multi-target prediction, from traditional supervised learning to modern graph-based and multi-task learning frameworks. We highlight real-world applications in oncology, central nervous system disorders, and drug repurposing, showcasing the translational potential of ML in systems pharmacology. Major challenges are discussed, such as data sparsity, lack of interpretability, limited generalizability, and integration into experimental workflows. We also address ethical and regulatory considerations surrounding model transparency, fairness, and reproducibility. Looking forward, we explore promising directions such as generative modeling, federated learning, and patient-specific therapy design. Together, these advances point toward a future of precision polypharmacology driven by biologically informed and interpretable ML models. This review aims to provide researchers and practitioners with a roadmap for leveraging ML in the development of safer and more effective multi-target therapeutics.

Graphical Abstract

1. Introduction

In recent years, the paradigm of drug discovery has been shifting from the traditional “one drug, one target” approach toward a more holistic and systems-level strategy—multi-target drug discovery [1,2]. This transformation is driven by the growing recognition that complex diseases such as cancer, neurodegenerative disorders, and metabolic syndromes often involve dysregulation of multiple genes, proteins, and pathways [3]. As a result, modulating a single molecular target may offer limited therapeutic benefit, whereas a well-designed multi-target intervention can improve efficacy, reduce resistance, and enhance safety profiles. This concept aligns with the principles of systems pharmacology, which integrates network biology, pharmacokinetics/pharmacodynamics (PK/PD), and computational modeling to understand drug action at the systems level [4].
Multi-target drugs are strategically designed to interact with a pre-defined set of molecular targets to achieve a synergistic therapeutic effect for complex diseases. This approach, known as rational polypharmacology, is a deliberate strategy to address the multifactorial nature of diseases [5,6]. In contrast, promiscuous drugs are compounds that exhibit a lack of specificity, binding to a wide range of unintended targets, which often leads to off-target effects and toxicity. While both types of drugs interact with multiple targets, the key distinction lies in the intentionality and specificity of their design. A multi-target drug’s target spectrum is carefully selected to contribute to the desired therapeutic outcome, whereas a promiscuous drug’s binding is often broad and unselective, potentially including many irrelevant or undesirable targets. Therefore, while a multi-target drug is by nature a ‘promiscuous’ binder, the term ‘promiscuous’ is often used pejoratively to denote a lack of specificity, whereas ‘multi-target’ implies a deliberate and beneficial polypharmacology [7].
However, identifying effective multi-target drug candidates remains a major challenge. The combinatorial explosion of potential drug-target interactions (DTI), the complexity of biological networks, and the limitations of high-throughput experimental screening demand scalable, accurate, and intelligent computational solutions [8]. In this context, machine learning (ML) has emerged as a powerful toolkit for modeling complex, nonlinear relationships inherent in biological systems [9,10]. By learning from diverse data sources—such as molecular structures, omics profiles, protein interactions, and clinical outcomes—ML algorithms can prioritize promising drug-target pairs, predict off-target effects, and even propose novel compounds with desirable polypharmacological profiles [11,12,13].
The integration of ML into multi-target drug discovery has opened new opportunities for understanding and optimizing complex therapeutic strategies, but it has also introduced significant challenges in data representation, model generalizability, and clinical applicability [14,15,16]. Classical ML models, such as support vector machines (SVMs), random forests (RFs), and logistic regression, have long demonstrated utility in tasks like predicting DTI, adverse effects, and pharmacokinetic profiles [17]. These models benefit from interpretability and robustness when trained on curated datasets. However, the growing volume and complexity of biomedical data have spurred the adoption of more sophisticated deep learning (DL) architectures [18,19]. In particular, graph neural networks (GNNs) excel at learning from molecular graphs and biological networks, while transformer-based models are increasingly leveraged to capture sequential, contextual, and multimodal biological information [18,20]. These approaches allow for the integration of chemical structure, target profiles, gene expression, and clinical phenotypes into unified predictive frameworks. Furthermore, the incorporation of systems pharmacology principles enables ML models to go beyond molecule-level predictions by considering the effects of drugs across pathways, tissues, and disease networks, facilitating a more holistic view of therapeutic efficacy and safety [21].
This review provides a comprehensive overview of how ML methods are applied to multi-target drug discovery, with an emphasis on their integration into systems pharmacology. We begin by outlining the biological rationale, then explore various ML techniques for feature representation, model construction, and network-based analysis. Key applications and case studies will be discussed to highlight their real-world implications. Finally, we address current challenges and propose future directions to guide research at the intersection of AI and multi-target pharmacology. The organizational framework is presented in Figure 1.

2. Fundamentals of Multi-Target Drug Discovery

Traditional drug discovery often follows a ‘one-drug, one-target’ paradigm, but this approach has limitations when addressing complex diseases. The evolution from this classical view to a systems pharmacology perspective is illustrated in Figure 2, which highlights the shift from single-target to multi-target and network-based therapeutic strategies.
The traditional paradigm of drug discovery has long focused on developing selective agents that modulate a single target associated with a specific disease [22]. While this approach has achieved success in certain therapeutic areas, it often falls short when applied to complex, multifactorial diseases such as cancer, neurodegenerative disorders, cardiovascular disease, and autoimmune conditions [23]. These diseases typically involve intricate biological networks with multiple dysregulated genes, signaling pathways, and feedback loops [24]. Consequently, interventions targeting a single node in such networks may lead to suboptimal efficacy, rapid resistance development, or unintended compensatory mechanisms. Multi-target drug discovery, or polypharmacology, offers a promising alternative by aiming to simultaneously modulate multiple targets involved in disease progression [25]. This strategy can produce synergistic therapeutic effects, enhance efficacy, and improve safety by reducing the doses required for each target [26]. In oncology, for instance, multi-kinase inhibitors have demonstrated improved clinical outcomes by blocking redundant signaling pathways that contribute to tumor survival [27]. In neurodegenerative diseases, addressing both amyloid accumulation and neuroinflammation through dual-target mechanisms holds greater promise than targeting either pathway alone [28,29]. Moreover, the concept of “network pharmacology” emphasizes that diseases are not the result of a single gene malfunction but rather the perturbation of interconnected networks [30]. From this perspective, therapeutic strategies should aim to restore network stability rather than simply block an individual target [31]. This system-level understanding forms the biological rationale for designing drugs that act on multiple molecular entities in a coordinated manner.
Despite the appeal of multi-target drug discovery, conventional experimental methods face significant limitations [23]. A major challenge lies in the combinatorial explosion of possible target sets and compound-target interactions [26]. With thousands of potential targets and millions of chemical compounds, the search space for discovering effective multi-target combinations becomes intractable using brute-force experimental techniques alone. Furthermore, traditional computational approaches—such as molecular docking or ligand-based virtual screening—often rely on predefined assumptions and simplified representations of molecular interactions [32,33]. Given these limitations, there is a pressing need for more sophisticated, data-driven approaches that can navigate the high-dimensional, nonlinear space of drug-target-disease interactions.

3. Machine Learning Techniques for Multi-Target Prediction

The complex and nonlinear nature of multi-target drug discovery requires computational methods that can efficiently model interactions across diverse chemical and biological spaces. ML has become a powerful approach to address these challenges, offering the flexibility to integrate heterogeneous data, learn hidden patterns, and make predictions at scale [34]. Figure 3 provides a visual overview of several representative pipelines, ranging from classical feature-based methods to modern DL architectures that leverage graph-based and attention mechanisms for more sophisticated predictions. This section outlines the key components of ML systems used for multi-target prediction, including data representations, classical and DL models, and network-based systems pharmacology frameworks.

3.1. Data Sources and Feature Representations

Effective ML for multi-target drug discovery relies heavily on rich, well-structured data representations derived from diverse biological and chemical domains [35]. Drug molecules can be encoded using a variety of representations, including molecular fingerprints (e.g., ECFP), SMILES strings, handcrafted molecular descriptors, and graph-based encodings that preserve structural topology [36,37]. On the other hand, targets—typically proteins—can be represented by their amino acid sequences, structural conformations, or contextual positions in protein–protein interaction (PPI) networks [38,39,40]. Modern embedding techniques such as those from pre-trained protein language models (e.g., ESM, ProtBERT) [41] and graph-based node embedding algorithms (e.g., DeepWalk, node2vec) [42] enable transformation of these entities into vectorized forms suitable for ML. Interaction data such as drug-target binding affinities or multi-label activity profiles are often collected from databases like DrugBank, ChEMBL, BindingDB, and STITCH [43]. A more comprehensive list of these and other key data sources is provided in Table 1. A central challenge is integrating such heterogeneous features into a unified learning framework [44], often addressed via feature fusion, co-embedding strategies, or representation learning.

3.2. Classical Machine Learning Models

Classical ML algorithms have laid the groundwork for multi-target drug discovery by offering scalable and interpretable tools for prediction. SVMs and RFs are widely adopted due to their strong performance in high-dimensional settings and robustness to overfitting [46,47]. These models have been used for both binary classification and regression tasks in DTI prediction. Other models such as k-nearest neighbors (k-NN) and naïve Bayes (NB) classifiers offer simplicity and speed, although they may lack the capacity to model complex relationships [48]. Multi-label classification strategies, including binary relevance, classifier chains, and label powerset transformations, have been employed to model drugs acting on multiple targets simultaneously [49]. However, classical models generally require extensive feature engineering and may not capture the higher-order dependencies found in biological systems, limiting their performance in highly nonlinear and interconnected pharmacological landscapes.

3.3. Deep Learning and Representation Learning

DL approaches have significantly advanced the capabilities of multi-target drug prediction by enabling automatic feature extraction and modeling of complex nonlinearities [50,51]. Multi-task learning (MTL) frameworks are particularly suited for multi-target settings, as they learn shared representations across related prediction tasks [52]. GNNs [53], including variants like graph convolutional networks (GCNs), graph attention networks (GATs), and GraphSAGE [54,55], have proven effective at modeling molecular graphs and predicting interactions by capturing rich spatial and relational information. A key architectural enhancement, residual GNNs, addresses the challenge of vanishing gradients in deep networks. By adding “skip connections” that allow information to bypass one or more layers, residual GNNs enable the training of much deeper models, facilitating the capture of complex, long-range dependencies within a molecule’s topological structure without losing crucial information from initial layers. This makes them particularly suitable for representing large and intricate chemical compounds [56]. Transformer-based models [57,58], originally developed for language modeling, are now applied to SMILES strings and protein sequences, with models like ChemBERTa [59], SMILES-BERT [60], and ProtTrans [61] demonstrating strong generalization in limited data scenarios. The core innovation of the Transformer is the self-attention mechanism. Unlike recurrent neural networks (RNNs) that process sequences sequentially, the attention mechanism allows the model to weigh the importance of all amino acids in a protein sequence simultaneously, regardless of their position. This parallel processing not only significantly improves computational efficiency but, more critically, allows the model to directly capture long-range dependencies and interactions between distant amino acids in a protein’s primary structure, which are often vital for its function and 3D folding. This capability makes Transformers a superior alternative for protein sequence representation compared to traditional sequential models [62]. Additionally, more refined multi-head attention-based models have been developed specifically for drug repurposing. These models leverage large-scale biological datasets to predict drug–disease relationships and demonstrate robust generalization capabilities, with some of their predictions indirectly confirmed by clinical trials [63]. Furthermore, unsupervised techniques such as autoencoder (AE), variational autoencoder (VAE) [64], and contrastive learning approaches allow the construction of latent representations even when labeled data are sparse [65]. Despite their predictive power, deep models often suffer from interpretability issues [66], raising concerns in clinical and regulatory settings.
The advent of generative artificial intelligence (AI) has revolutionized the paradigm of multi-target drug discovery by shifting from a traditional ‘screen-and-select’ approach to a ‘design-and-generate’ one. These models, trained on vast datasets of chemical structures and their associated biological activities, can learn the underlying chemical and pharmacological rules to create entirely novel molecules from scratch, a process known as de novo design [14,67]. A key advantage of generative AI in this context is its ability to explore the immense chemical space more effectively than traditional methods. Models such as VAEs, generative adversarial networks (GANs) [68], and diffusion models [69] are used to produce molecular structures with specific, pre-defined properties, including the desired multi-target profiles. These models are particularly suited for polypharmacology because they can be optimized to generate compounds that simultaneously satisfy multiple objectives, such as binding affinity to several targets, drug-likeness, and synthetic accessibility [70]. For instance, recent studies have demonstrated the use of generative models to design multi-target ligands for complex diseases, showcasing their potential to accelerate the development of more effective and safer therapeutics [71,72].
In summary, each of the discussed approaches offers distinct trade-offs in multi-target drug discovery. Classical ML models (Section 3.2) excel in tasks with limited, well-defined data, offering high interpretability and lower computational costs [73]. However, their reliance on manual feature engineering and limited capacity to model complex, hierarchical relationships hinder their scalability for large, high-dimensional datasets. In contrast, DL methods (Section 3.3), particularly GNNs and Transformers, demonstrate superior performance and scalability by automatically learning complex patterns from vast, heterogeneous data [74]. While DL excels in predictive power, it often suffers from poor interpretability, a critical limitation in a field requiring high-stakes, explainable decisions. The emerging systems pharmacology approaches and hybrid models (Section 3.4) bridge this gap by integrating diverse data sources and network contexts. They offer a promising path to not only improve predictive accuracy but also enhance the biological relevance and interpretability of results, making them highly suitable for complex, systems-level therapeutic tasks like multi-target drug repurposing and de novo design [5].

3.4. Network-Based and Systems Pharmacology Approaches

ML in systems pharmacology often leverages the structure of biological networks to contextualize DTI [75,76]. Heterogeneous networks, which represent drugs, proteins, diseases, and pathways as interconnected nodes, offer a rich substrate for prediction via link prediction or network completion algorithms [77]. Embedding methods such as node2vec [78], metapath2vec [79], and graph AE allow transformation of these complex networks into low-dimensional feature spaces while preserving topological and semantic relationships. In addition, network propagation [80] and diffusion algorithms [81] simulate how drug perturbations influence biological systems, identifying synergistic multi-target profiles and compensatory pathways. Knowledge graphs further extend these capabilities by encoding semantic relations among biomedical entities [82]; when combined with embedding methods like TransE or GNN-based link predictors [83], they support reasoning over complex associations and contribute to explainable predictions [78]. These systems-level approaches provide the necessary context for understanding polypharmacology beyond molecular interactions and support network-informed therapeutic design. These methods can identify not just potential drug candidates but also the optimal network nodes to modulate for desired therapeutic outcomes.
A key trend is to integrate factors beyond direct drug-target binding, such as pharmacokinetics and the influence of the gut microbiota (GM). A recent study using an animal model showed that the GM can significantly alter the pharmacokinetics of drugs, thereby affecting their efficacy and potential for drug interactions [84]. This highlights the importance of a systems-level approach that considers not only molecular interactions but also physiological and microbial factors to provide a more complete picture of drug action.
The ability of knowledge graphs to encode vast, heterogeneous biomedical data makes them particularly powerful for novel target discovery and drug repurposing, which are essential facets of multi-target therapy. Recent advancements have led to the development of sophisticated graph foundation models that perform zero-shot drug repurposing, identifying therapeutic candidates for diseases with limited or no existing treatments. A prime example is TxGNN, a graph neural network model trained on a comprehensive medical knowledge graph [85]. TxGNN uses a metric learning module to rank potential drug indications and contraindications, showcasing a significant improvement in prediction accuracy over conventional methods. A key feature of such models is their interpretability, facilitated by modules like TxGNN’s Explainer. This component provides transparent, multi-hop knowledge paths that allow human experts to investigate and validate the model’s predictions, thereby building trust and bridging the gap between computational results and clinical decision-making. These innovations highlight the potential of knowledge graphs not only for predicting known DTI but also for discovering entirely new therapeutic connections within the complex landscape of diseases. Beyond the molecular level, the scope of multi-target therapy extends to encompass the systemic, multi-organ nature of chronic diseases. This holistic perspective is crucial because many complex conditions, such as metabolic or neurodegenerative disorders, involve an intricate web of malfunctioning organs and disrupted inter-organ communication. A recent concept, the locked-state model (LoSM), illustrates how these pathological states can become ‘locked’ due to body-wide, memory-like properties resulting from inter-organ crosstalk [86]. Deciphering these complex, multi-organ locked states present a significant opportunity for systems pharmacology. Applying ML and systems biology at this multi-scale level enables the identification of therapeutic strategies that can dismantle pathological states by targeting key nodes within these complex biological systems. This approach not only facilitates the discovery of optimal multi-target profiles but also helps in designing interventions that can modulate an entire system rather than just a single pathway. These advancements highlight a new frontier in multi-target therapy, where the goal is to develop treatments that restore healthy inter-organ communication and unlock chronic pathological states.

4. Applications and Case Studies

ML-based strategies for multi-target drug discovery have transitioned from theoretical frameworks to practical applications across a wide spectrum of therapeutic areas. These applications demonstrate the feasibility of ML techniques and highlight their capacity to uncover novel DTI, optimize combination therapies, and accelerate drug repurposing. This section presents representative case studies that illustrate how ML is being applied to real-world drug discovery challenges in oncology, neurodegenerative diseases, infectious diseases, and personalized therapy. As shown in Figure 4, these diverse applications reveal a common theme: ML models are evolving to integrate different data types and address the unique, multifaceted challenges of each therapeutic area, paving the way for a new era of precision polypharmacology.

4.1. Oncology: Targeting Redundant and Synergistic Pathways

As highlighted in Figure 4, cancer presents a primary area for ML-guided multi-target drug discovery due to its intrinsic biological complexity, including pathway redundancy, tumor heterogeneity, and adaptive resistance mechanisms [87,88]. The use of multi-target therapeutics in oncology is crucial for overcoming resistance and achieving synergistic effects. Table 2 provides a summary of representative studies that utilize ML for drug combination and synergy prediction in cancer treatment. ML models have been applied to identify kinase inhibitors capable of targeting multiple oncogenic pathways simultaneously. For instance, DL models integrating molecular graph data and transcriptomic signatures have been used to predict dual or triple kinase inhibitors with improved efficacy across different cancer cell lines [89,90]. In another example, a graph-based ML framework was used to identify multitarget compounds capable of co-inhibiting PI3K, mTOR, and EGFR—three pathways frequently co-activated in lung and breast cancers [91,92,93]. These predictions were validated with in vitro and xenograft studies, underscoring the translational potential of ML-guided approaches. Additionally, ML-assisted drug combination prediction has emerged as a complementary strategy, where models forecast synergistic drug pairs based on cell line–specific omics data and pathway activity scores, enabling more effective multi-target treatment regimens [94].

4.2. Neurodegenerative Diseases: Tackling Multifactorial Pathogenesis

Neurodegenerative diseases such as Alzheimer’s and Parkinson’s disease (AD and PD) involve multiple pathogenic processes, including protein aggregation, mitochondrial dysfunction, oxidative stress, and neuroinflammation [118]. Given the multifactorial pathogenesis of neurodegenerative disorders, multi-target strategies are particularly well-suited for their treatment. A summary of key studies that have employed ML for multi-target drug discovery in this area is provided in Table 3. Conventional single-target strategies have failed to deliver effective disease-modifying therapies, prompting a shift toward systems-level approaches. ML models have been trained on chemical features, transcriptomic responses, and protein interaction data to predict compounds that simultaneously modulate amyloid-beta aggregation and neuroinflammatory pathways [119,120,121]. For example, multi-task neural networks have been employed to predict ligands capable of dual inhibition of BACE1 and GSK-3β, both implicated in AD’s progression [121,122]. Moreover, knowledge graph–based embedding models have facilitated the identification of repositioned drugs that act on multiple neurodegeneration-associated targets [123,124], such as donepezil analogs with additional antioxidant or anti-inflammatory properties [125,126].
Antimicrobial resistance (AMR) presents a growing public health crisis, where multi-target therapies offer a way to prevent rapid resistance evolution [23]. The growing threat of AMR has necessitated the development of new therapeutic strategies. Table 4 highlights several important studies where ML has been applied to combat infectious diseases and predict resistance patterns. ML approaches have been used to identify multi-target antibiotics that interfere with essential bacterial processes such as DNA replication, protein synthesis, and cell wall biosynthesis [131,132]. For instance, supervised learning models trained on pathogen-specific compound libraries have predicted antibiotics that simultaneously bind to DNA gyrase and topoisomerase IV—an established strategy to overcome resistance in Gram-negative bacteria [133,134]. In the context of viral infections like COVID-19, network-based ML models have been used to repurpose existing drugs with multitarget activities against both viral proteins and host factors involved in viral entry and replication [135]. These methods offer a scalable means to rapidly identify broad-spectrum antivirals with polypharmacological profiles.
One of the most promising applications of ML in multi-target discovery lies in drug repurposing and personalized therapy [87,156]. Integrative ML frameworks that incorporate patient-specific omics profiles, electronic health records (EHRs), and pharmacogenomic data are now being used to predict individualized drug combinations. For example, ML models have been developed to stratify patients based on molecular subtypes and predict the optimal multi-target regimen that addresses their unique pathway dysregulations [23,157]. In oncology, this approach has led to patient-specific predictions of kinase inhibitor combinations with minimal off-target toxicity [158]. In another instance, reinforcement learning models have been applied to iteratively optimize drug dosing schedules and combinations for rare diseases, using patient-level simulations and prior clinical knowledge [159,160]. This move toward precision polypharmacology is evident in studies that leverage computational methods for drug repurposing and developing personalized strategies. For example, one study applied a DL model based on BERT for text classification to a benchmark dataset of Traditional Chinese Medicine (TCM) clinical records [161]. The research aimed to classify TCM clinical records into five main disease categories, with the BERT-based method achieving state-of-the-art accuracy and F1 scores. In another instance, the PanGPCR system was developed to predict multiple GPCR targets and repurposing potential for GPCR-targeting drugs. This system uses molecular docking to predict binding affinities and targets. PanGPCR can identify a compound’s potential GPCR targets and repurposing potential by generating a ranked list from docking studies, which can be linked to side effect data from the SIDER database [162]. Such personalized systems pharmacology frameworks represent a significant step toward precision polypharmacology, where treatment strategies are tailored not only to disease mechanisms but also to the molecular context of individual patients.

5. Challenges and Future Perspectives

Despite the promising potential of ML in multi-target drug discovery, significant challenges remain. A primary obstacle is the lack of high-quality, large-scale datasets, which are often sparse, heterogeneous, and imbalanced, leading to issues like overfitting and poor generalizability [163,164,165]. Specifically, dataset biases are a significant concern, as publicly available databases often overrepresent well-studied compounds and targets. This can result in models that perform well on existing data but fail to generalize to novel chemical spaces or underrepresented target families. Similarly, imbalanced class distributions are a pervasive issue, as drug activity datasets contain a vast majority of inactive compounds. This can cause models to be biased toward the majority negative class, making them insensitive to the rare but crucial active compounds. The problem is further compounded by the challenge of missing data, where incomplete activity profiles or heterogeneous data formats require sophisticated imputation or specialized machine learning methods to build a unified, robust training set. Furthermore, the “black-box” nature of complex models, such as deep learning, hinders interpretability, making it difficult for chemists and regulatory agencies to understand the biological reasoning behind predictions [72,166]. Models also struggle to generalize across different biological contexts and disease states. A final challenge is integrating these computational predictions into experimental and clinical pipelines, as well as addressing ethical and regulatory concerns surrounding data privacy, algorithmic bias, and model reproducibility [73].
The future of multi-target drug discovery will likely be shaped by synergistic advances that address current limitations. Key areas include developing standardized, high-quality datasets and more interpretable ML models to provide clearer biological insights [74]. Advances in transfer learning and domain adaptation will also improve model generalizability across diverse biological contexts [167,168]. The field is moving toward a more collaborative approach, bridging the gap between computational and experimental pipelines. Looking ahead, the emergence of generative AI, federated learning, and the integration of patient-level omics data are expected to accelerate the development of personalized, multi-target therapies, moving us toward a future of precision polypharmacology [169].

6. Conclusions

The integration of machine learning into multi-target drug discovery is a transformative shift in pharmacology, moving beyond traditional single-target methods to better address complex diseases like cancer and neurodegeneration. This approach leverages powerful techniques such as deep learning and graph neural networks to enable high-throughput analysis and predictive modeling. While promising applications in cancer therapy and drug repurposing have been demonstrated, significant challenges remain, including data sparsity and a lack of model interpretability. The future of this field depends on advances in data integration, transfer learning, and interdisciplinary collaboration, with generative AI and patient-level omics data poised to further enhance the development of safer, more personalized therapeutic interventions.

Author Contributions

Conceptualization, X.B. and C.L.; methodology, Y.W. and J.W.; software, Y.W. and J.W.; validation, J.W. and C.L.; data curation, X.B.; writing—original draft preparation, X.B. and Y.W.; writing—review and editing, C.L.; visualization, Y.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Research Project of Yan’an University, grant number YAU202512552.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

PK: pharmacokinetics; PD: pharmacodynamics; DTI: drug-target interactions; ML: machine learning; SVMs: support vector machines; RFs: random forests; DL: deep learning; GNNs: graph neural networks; GCNs: graph convolutional networks; GATs: graph attention networks; MLP: multi-layer perceptron; PPI: protein–protein interaction; NCI: National Cancer Institute; k-NN: k-nearest neighbors; NB: naïve Bayes; MTL: multi-task learning; RNNs: recurrent neural networks; AE: autoencoder; VAE: variational autoencoder; AI: artificial intelligence; GANs: generative adversarial networks; GM: gut microbiota; LoSM: locked-state model; DNN: deep neural network; AUC: area under the curve; ROC: receiver operating characteristic curve; TPR: true positive rate; DDS: drug–drug synergy; LRGA: low-rank global attention; MGAE: multi-channel graph autoencoder; MSE: mean squared error; IDA: independent drug action; AD: Alzheimer’s disease; PD: Parkinson’s disease; DTA: drug-target affinity; AMR: antimicrobial resistance; ARG: antibiotic resistance genes; MIC: minimum inhibitory concentration; AMPs: antimicrobial peptides; WGS: whole-genome sequence; EHRs: electronic health records; TCM: Traditional Chinese Medicine.

References

  1. Lavecchia, A.; Cerchia, C. In Silico Methods to Address Polypharmacology: Current Status, Applications and Future Perspectives. Drug Discov. Today 2016, 21, 288–298. [Google Scholar] [CrossRef]
  2. Zhou, W.; Wang, Y.; Lu, A.; Zhang, G. Systems Pharmacology in Small Molecular Drug Discovery. Int. J. Mol. Sci. 2016, 17, 246. [Google Scholar] [CrossRef]
  3. Pérez-Nueno, V.I. Using Quantitative Systems Pharmacology for Novel Drug Discovery. Expert Opin. Drug Discov. 2015, 10, 1315–1331. [Google Scholar] [CrossRef]
  4. Li, L.; Yang, L.; Yang, L.; He, C.; He, Y.; Chen, L.; Dong, Q.; Zhang, H.; Chen, S.; Li, P. Network Pharmacology: A Bright Guiding Light on the Way to Explore the Personalized Precise Medication of Traditional Chinese Medicine. Chin. Med. 2023, 18, 146. [Google Scholar] [CrossRef] [PubMed]
  5. Ryszkiewicz, P.; Malinowska, B.; Schlicker, E. Polypharmacology: New Drugs in 2023–2024. Pharmacol. Rep. 2025, 77, 543–560. [Google Scholar] [CrossRef] [PubMed]
  6. Stefan, S.M.; Rafehi, M. Medicinal Polypharmacology-a Scientific Glossary of Terminology and Concepts. Front. Pharmacol. 2024, 15, 1419110. [Google Scholar] [CrossRef]
  7. VanDongen, A.M. Drug Promiscuity: Problems and Promises. bioRxiv 2023. [Google Scholar] [CrossRef]
  8. Abdelsayed, M. AI-Driven Polypharmacology in Small-Molecule Drug Discovery. Int. J. Mol. Sci. 2025, 26, 6996. [Google Scholar] [CrossRef]
  9. Liu, K.; Chen, X.; Ren, Y.; Liu, C.; Lv, T.; Liu, Y.; Zhang, Y. Multi-Target-Based Polypharmacology Prediction (mTPP): An Approach Using Virtual Screening and Machine Learning for Multi-Target Drug Discovery. Chem.-Biol. Interact. 2022, 368, 110239. [Google Scholar] [CrossRef] [PubMed]
  10. Kleandrova, V.V.; Ds Cordeiro, M.N.; Speck-Planche, A. Current In Silico Methods for Multi-Target Drug Discovery in Early Anticancer Research: The Rise of the Perturbation-Theory Machine Learning Approach. Future Med. Chem. 2023, 15, 1647–1650. [Google Scholar] [CrossRef]
  11. Cichońska, A.; Ravikumar, B.; Rahman, R. AI for Targeted Polypharmacology: The next Frontier in Drug Discovery. Curr. Opin. Struct. Biol. 2024, 84, 102771. [Google Scholar] [CrossRef]
  12. Mukaidaisi, M.; Ahmed, M.; Grantham, K.; Al-Jumaily, A.; Dedhar, S.; Organ, M.; Tchagang, A.; Hou, J.; Ahmed, S.E.; Dividino, R.; et al. “Several Birds with One Stone”: Exploring the Potential of AI Methods for Multi-Target Drug Design. Mol. Divers. 2025, 29, 3023–3039. [Google Scholar] [CrossRef]
  13. Isigkeit, L.; Hörmann, T.; Schallmayer, E.; Scholz, K.; Lillich, F.F.; Ehrler, J.H.M.; Hufnagel, B.; Büchner, J.; Marschner, J.A.; Pabel, J.; et al. Automated Design of Multi-Target Ligands by Generative Deep Learning. Nat. Commun. 2024, 15, 7946. [Google Scholar] [CrossRef]
  14. Ocana, A.; Pandiella, A.; Privat, C.; Bravo, I.; Luengo-Oroz, M.; Amir, E.; Gyorffy, B. Integrating Artificial Intelligence in Drug Discovery and Early Drug Development: A Transformative Approach. Biomark. Res. 2025, 13, 45. [Google Scholar] [CrossRef]
  15. Ogbonna, U.E.; Ofoezie, E.F.; Babalola, O.O.; Ottu, P.O.; Ogbonna, C.A.; Olisakwe, S.; George, T.E.; Babarinde, S.; Omaba, J.O.; Chukwuemeka, C.G.; et al. Advances in Machine Learning for Optimizing Pharmaceutical Drug Discovery. Curr. Proteom. 2025, 22, 100015. [Google Scholar] [CrossRef]
  16. Bajorath, J. Explainable Machine Learning for Medicinal Chemistry: Exploring Multi-Target Compounds. Future Med. Chem. 2022, 14, 1171–1173. [Google Scholar] [CrossRef] [PubMed]
  17. Serrano, D.R.; Luciano, F.C.; Anaya, B.J.; Ongoren, B.; Kara, A.; Molina, G.; Ramirez, B.I.; Sánchez-Guirales, S.A.; Simon, J.A.; Tomietto, G.; et al. Artificial Intelligence (AI) Applications in Drug Discovery and Drug Delivery: Revolutionizing Personalized Medicine. Pharmaceutics 2024, 16, 1328. [Google Scholar] [CrossRef] [PubMed]
  18. Kandhare, P.; Kurlekar, M.; Deshpande, T.; Pawar, A. A Review on Revolutionizing Healthcare Technologies with AI and ML Applications in Pharmaceutical Sciences. Drugs Drug Candidates 2025, 4, 9. [Google Scholar] [CrossRef]
  19. Talukder, M.A.; Kazi, M.; Alazab, A. Predicting Drug-Target Interactions Using Machine Learning with Improved Data Balancing and Feature Engineering. Sci. Rep. 2025, 15, 19495. [Google Scholar] [CrossRef]
  20. Ali, N.; Hanif, N.; Khan, H.A.; Waseem, M.A.; Saeed, A.; Zakir, S.; Khan, A.; Aamir, M.; Ali, A.; Ali, A.; et al. Deep Learning and Artificial Intelligence for Drug Discovery, Application, Challenge, and Future Perspectives. Discov. Appl. Sci. 2025, 7, 533. [Google Scholar] [CrossRef]
  21. Wan, Z.; Sun, X.; Li, Y.; Chu, T.; Hao, X.; Cao, Y.; Zhang, P. Applications of Artificial Intelligence in Drug Repurposing. Adv. Sci. 2025, 12, 2411325. [Google Scholar] [CrossRef]
  22. Hughes, J.; Rees, S.; Kalindjian, S.; Philpott, K. Principles of Early Drug Discovery. Br. J. Pharmacol. 2011, 162, 1239–1249. [Google Scholar] [CrossRef]
  23. Makhoba, X.H.; Viegas, C., Jr.; Mosa, R.A.; Viegas, F.P.D.; Pooe, O.J. Potential Impact of the Multi-Target Drug Approach in the Treatment of Some Complex Diseases. Drug Des. Dev. Ther. 2020, 14, 3235–3249. [Google Scholar] [CrossRef]
  24. Li, Q.; Geng, S.; Luo, H.; Wang, W.; Mo, Y.-Q.; Luo, Q.; Wang, L.; Song, G.-B.; Sheng, J.-P.; Xu, B. Signaling Pathways Involved in Colorectal Cancer: Pathogenesis and Targeted Therapy. Signal Transduct. Target. Ther. 2024, 9, 266. [Google Scholar] [CrossRef]
  25. Chaudhari, R.; Tan, Z.; Huang, B.; Zhang, S. Computational Polypharmacology: A New Paradigm for Drug Discovery. Expert Opin. Drug Discov. 2017, 12, 279–291. [Google Scholar] [CrossRef] [PubMed]
  26. Talevi, A. Multi-Target Pharmacology: Possibilities and Limitations of the “Skeleton Key Approach” from a Medicinal Chemist Perspective. Front. Pharmacol. 2015, 6, 205. [Google Scholar] [CrossRef]
  27. Yesilkanal, A.E.; Johnson, G.L.; Ramos, A.F.; Rosner, M.R. New strategies for targeting kinase networks in cancer. J. Biol. Chem. 2021, 297, 101128. [Google Scholar] [CrossRef]
  28. Leri, M.; Vasarri, M. Advancing Neuropharmacology and Neurodegenerative Disease Therapy: Bridging Gaps and Paving New Pathways. Pharmaceuticals 2025, 18, 606. [Google Scholar] [CrossRef]
  29. Iqbal, D.; Rehman, M.T.; Alajmi, M.F.; Alsaweed, M.; Jamal, Q.M.S.; Alasiry, S.M.; Albaker, A.B.; Hamed, M.; Kamal, M.; Albadrani, H.M. Multitargeted Virtual Screening and Molecular Simulation of Natural Product-like Compounds against GSK3β, NMDA-Receptor, and BACE-1 for the Management of Alzheimer’s Disease. Pharmaceuticals 2023, 16, 622. [Google Scholar] [CrossRef]
  30. Chandran, U.; Mehendale, N.; Patil, S.; Chaguturu, R.; Patwardhan, B. Network Pharmacology. In Innovative Approaches in Drug Discovery; Elsevier: Amsterdam, The Netherlands, 2017; pp. 127–164. [Google Scholar] [CrossRef]
  31. Hopkins, A.L. Network Pharmacology: The next Paradigm in Drug Discovery. Nat. Chem. Biol. 2008, 4, 682–690. [Google Scholar] [CrossRef]
  32. Cavasotto, C.N.; Orry, A.J.W. Ligand Docking and Structure-Based Virtual Screening in Drug Discovery. Curr. Top. Med. Chem. 2007, 7, 1006–1014. [Google Scholar] [CrossRef]
  33. Pal, S.; Kumar, V.; Kundu, B.; Bhattacharya, D.; Preethy, N.; Reddy, M.P.; Talukdar, A. Ligand-Based Pharmacophore Modeling, Virtual Screening and Molecular Docking Studies for Discovery of Potential Topoisomerase I Inhibitors. Comput. Struct. Biotechnol. J. 2019, 17, 291–310. [Google Scholar] [CrossRef]
  34. Rehman, A.U.; Li, M.; Wu, B.; Ali, Y.; Rasheed, S.; Shaheen, S.; Liu, X.; Luo, R.; Zhang, J. Role of Artificial Intelligence in Revolutionizing Drug Discovery. Fundam. Res. 2025, 5, 1273–1287. [Google Scholar] [CrossRef]
  35. Wigh, D.S.; Goodman, J.M.; Lapkin, A.A. A Review of Molecular Representation in the Age of Machine Learning. WIREs Comput. Mol. Sci. 2022, 12, e1603. [Google Scholar] [CrossRef]
  36. Li, Z.; Jiang, M.; Wang, S.; Zhang, S. Deep Learning Methods for Molecular Representation and Property Prediction. Drug Discov. Today 2022, 27, 103373. [Google Scholar] [CrossRef]
  37. Xu, Z.; Wang, S.; Zhu, F.; Huang, J. Seq2seq Fingerprint: An Unsupervised Deep Molecular Embedding for Drug Discovery. In Proceedings of the 8th ACM International Conference on Bioinformatics, Computational Biology, and Health Informatics, Boston, MA, USA, 20–23 August 2017; ACM: Boston, MA, USA, 2017; pp. 285–294. [Google Scholar]
  38. Xie, X.; Yu, T.; Li, X.; Zhang, N.; Foster, L.J.; Peng, C.; Huang, W.; He, G. Recent Advances in Targeting the “Undruggable” Proteins: From Drug Discovery to Clinical Trials. Signal Transduct. Target. Ther. 2023, 8, 335. [Google Scholar] [CrossRef]
  39. Lahti, J.L.; Tang, G.W.; Capriotti, E.; Liu, T.; Altman, R.B. Bioinformatics and Variability in Drug Response: A Protein Structural Perspective. J. R. Soc. Interface 2012, 9, 1409–1437. [Google Scholar] [CrossRef]
  40. Nero, T.L.; Parker, M.W.; Morton, C.J. Protein Structure and Computational Drug Discovery. Biochem. Soc. Trans. 2018, 46, 1367–1379. [Google Scholar] [CrossRef]
  41. Djeddi, W.E.; Hermi, K.; Ben Yahia, S.; Diallo, G. Advancing Drug–Target Interaction Prediction: A Comprehensive Graph-Based Approach Integrating Knowledge Graph Embedding and ProtBert Pretraining. BMC Bioinform. 2023, 24, 488. [Google Scholar] [CrossRef]
  42. Kim, M.; Baek, S.H.; Song, M. Relation Extraction for Biological Pathway Construction Using Node2vec. BMC Bioinform. 2018, 19, 206. [Google Scholar] [CrossRef]
  43. Ren, X.; Yan, C.-X.; Zhai, R.-X.; Xu, K.; Li, H.; Fu, X.-J. Comprehensive Survey of Target Prediction Web Servers for Traditional Chinese Medicine. Heliyon 2023, 9, e19151. [Google Scholar] [CrossRef] [PubMed]
  44. Zhang, Y.; Liu, C.; Liu, M.; Liu, T.; Lin, H.; Huang, C.-B.; Ning, L. Attention Is All You Need: Utilizing Attention in AI-Enabled Drug Discovery. Brief. Bioinform. 2023, 25, bbad467. [Google Scholar] [CrossRef]
  45. O’Neil, J.; Benita, Y.; Feldman, I.; Chenard, M.; Roberts, B.; Liu, Y.; Li, J.; Kral, A.; Lejnine, S.; Loboda, A.; et al. An Unbiased Oncology Compound Screen to Identify Novel Combination Strategies. Mol. Cancer Ther. 2016, 15, 1155–1162. [Google Scholar] [CrossRef]
  46. Rodríguez-Pérez, R.; Bajorath, J. Evolution of Support Vector Machine and Regression Modeling in Chemoinformatics and Drug Discovery. J. Comput. Aided Mol. Des. 2022, 36, 355–362. [Google Scholar] [CrossRef]
  47. Nayarisseri, A.; Khandelwal, R.; Tanwar, P.; Madhavi, M.; Sharma, D.; Thakur, G.; Speck-Planche, A.; Singh, S.K. Artificial Intelligence, Big Data and Machine Learning Approaches in Precision Medicine & Drug Discovery. Curr. Drug Targets 2021, 22, 631–655. [Google Scholar] [CrossRef]
  48. Indrajaya, D.; Setiawan, A.; Susanto, B. Comparison of K-Nearest Neighbor and Naive Bayes Methods for SNP Data Classification. MATRIK J. Manaj. Tek. Inform. Rekayasa Komput. 2022, 22, 149–164. [Google Scholar] [CrossRef]
  49. Bai, T.; Xie, J.; Liu, Y.; Liu, B. MMLmiRLocNet: miRNA Subcellular Localization Prediction Based on Multi-View Multi-Label Learning for Drug Design. IEEE J. Biomed. Health Inform. 2024, 1–9. [Google Scholar] [CrossRef]
  50. Das, P.; Sercu, T.; Wadhawan, K.; Padhi, I.; Gehrmann, S.; Cipcigan, F.; Chenthamarakshan, V.; Strobelt, H.; Dos Santos, C.; Chen, P.-Y.; et al. Accelerated Antimicrobial Discovery via Deep Generative Models and Molecular Dynamics Simulations. Nat. Biomed. Eng. 2021, 5, 613–623. [Google Scholar] [CrossRef]
  51. Liu, H.; Lin, X.; Hu, J.; Zhang, X. A Multi Target Drug Design Method Based on Target Protein Sequence and Feature Similarity. In Proceedings of the 2024 IEEE International Conference on Bioinformatics and Biomedicine (BIBM), Lisbon, Portugal, 3–6 December 2024; IEEE: Lisbon, Portugal, 2024; pp. 848–851. [Google Scholar]
  52. Born, J.; Manica, M. Trends in Deep Learning for Property-Driven Drug Design. Curr. Med. Chem. 2021, 28, 7862–7886. [Google Scholar] [CrossRef]
  53. Qi, G.; Zhang, A.; Xu, Z.; Li, Z.; Zeng, W.; Liu, X.; Ma, J.; Zheng, X.; Li, Z. Application of a Complex Network Modeling Approach to Explore the Material Basis and Mechanisms of Traditional Chinese Medicine: A Case Study of Xuefu Zhuyu Decoction for the Treatment of Two Types of Angina Pectoris. IEEE Access 2022, 10, 114103–114117. [Google Scholar] [CrossRef]
  54. Tang, X.; Lei, X.; Liu, L. A Multi-Modal Drug Target Affinity Prediction Based on Graph Features and Pre-Trained Sequence Embeddings. Interdiscip. Sci. Comput. Life Sci. 2025, 1–22. [Google Scholar] [CrossRef]
  55. Zeng, X.; Su, G.-P.; Du, W.-F.; Jiang, B.; Li, Y.; Yang, Z.-Z. Joint Fusion of Sequences and Structures of Drugs and Targets for Identifying Targets Based on Intra and Inter Cross-Attention Mechanisms. BMC Biol. 2025, 23, 168. [Google Scholar] [CrossRef]
  56. Zonyfar, C.; Njimbouom, S.N.; Mosalla, S.; Kim, J.-D. R2eGIN: Residual Reconstruction Enhanced Graph Isomorphism Network for Accurate Prediction of Poly (ADP-Ribose) Polymerase Inhibitors. Bioinform. Biol. Insights 2025, 19, 11779322251366087. [Google Scholar] [CrossRef]
  57. Ai, C.; Yang, H.; Liu, X.; Dong, R.; Ding, Y.; Guo, F. MTMol-GPT: De Novo Multi-Target Molecular Generation with Transformer-Based Generative Adversarial Imitation Learning. PLoS Comput. Biol. 2024, 20, e1012229. [Google Scholar] [CrossRef]
  58. Mao, J.; Wang, J.; Zeb, A.; Cho, K.-H.; Jin, H.; Kim, J.; Lee, O.; Wang, Y.; No, K.T. Transformer-Based Molecular Generative Model for Antiviral Drug Design. J. Chem. Inf. Model. 2024, 64, 2733–2745. [Google Scholar] [CrossRef]
  59. Wang, L.; Wang, S.; Yang, H.; Li, S.; Wang, X.; Zhou, Y.; Tian, S.; Liu, L.; Bai, F. Conformational Space Profiling Enhances Generic Molecular Representation for AI-Powered Ligand-Based Drug Discovery. Adv. Sci. 2024, 11, 2403998. [Google Scholar] [CrossRef]
  60. Zhang, Q.; Ding, K.; Lyv, T.; Wang, X.; Yin, Q.; Zhang, Y.; Yu, J.; Wang, Y.; Li, X.; Xiang, Z.; et al. Scientific Large Language Models: A Survey on Biological & Chemical Domains. arXiv 2024, arXiv:2401.14656. [Google Scholar] [CrossRef]
  61. Zhao, Y.; Xing, Y.; Zhang, Y.; Wang, Y.; Wan, M.; Yi, D.; Wu, C.; Li, S.; Xu, H.; Zhang, H.; et al. Evidential Deep Learning-Based Drug-Target Interaction Prediction. Nat. Commun. 2025, 16, 6915. [Google Scholar] [CrossRef] [PubMed]
  62. Ghosh, N.; Santoni, D.; Nawn, D.; Ottaviani, E.; Felici, G. A Comprehensive Review of Transformer-Based Language Models for Protein Sequence Analysis and Design. arXiv 2025, arXiv:2507.13646. [Google Scholar] [CrossRef]
  63. Feng, X.; Ma, Z.; Yu, C.; Xin, R. MRNDR: Multihead Attention-Based Recommendation Network for Drug Repurposing. J. Chem. Inf. Model. 2024, 64, 2654–2669. [Google Scholar] [CrossRef] [PubMed]
  64. Li, Y.; Liu, X.; Zhou, J.; Li, F.; Wang, Y.; Liu, Q. Artificial Intelligence in Traditional Chinese Medicine: Advances in Multi-Metabolite Multi-Target Interaction Modeling. Front. Pharmacol. 2025, 16, 1541509. [Google Scholar] [CrossRef]
  65. Vilalta-Mor, J.; Molina, A.; Varga, L.O.; Filella-Merce, I.; Guallar, V. Active Learning-Guided Seq2Seq Variational Autoencoder for Multi-Target Inhibitor Generation. arXiv 2025, arXiv:2506.15309. [Google Scholar] [CrossRef]
  66. Teng, Q.; Liu, Z.; Song, Y.; Han, K.; Lu, Y. A Survey on the Interpretability of Deep Learning in Medical Diagnosis. Multimed. Syst. 2022, 28, 2335–2355. [Google Scholar] [CrossRef]
  67. Gangwal, A.; Ansari, A.; Ahmad, I.; Azad, A.K.; Kumarasamy, V.; Subramaniyan, V.; Wong, L.S. Generative Artificial Intelligence in Drug Discovery: Basic Framework, Recent Advances, Challenges, and Opportunities. Front. Pharmacol. 2024, 15, 1331062. [Google Scholar] [CrossRef] [PubMed]
  68. Wang, S.; Lin, T.; Peng, T.; Xing, E.; Chen, S.; Kara, L.B.; Cheng, X. TopMT-GAN: A 3D Topology-Driven Generative Model for Efficient and Diverse Structure-Based Ligand Design. Chem. Sci. 2025, 16, 2796–2809. [Google Scholar] [CrossRef]
  69. Zheng, J.; Yi, H.-C.; You, Z.-H. Equivariant 3D-Conditional Diffusion Model for De Novo Drug Design. IEEE J. Biomed. Health Inform. 2025, 29, 1805–1816. [Google Scholar] [CrossRef]
  70. Fang, Y.; Pan, X.; Shen, H.-B. De Novo Drug Design by Iterative Multiobjective Deep Reinforcement Learning with Graph-Based Molecular Quality Assessment. Bioinformatics 2023, 39, btad157. [Google Scholar] [CrossRef] [PubMed]
  71. Abdelbaky, I.; Tayara, H.; Chong, K.T. Prediction of Kinase Inhibitors Binding Modes with Machine Learning and Reduced Descriptor Sets. Sci. Rep. 2021, 11, 706. [Google Scholar] [CrossRef] [PubMed]
  72. Feldmann, C.; Bajorath, J. Differentiating Inhibitors of Closely Related Protein Kinases with Single- or Multi-Target Activity via Explainable Machine Learning and Feature Analysis. Biomolecules 2022, 12, 557. [Google Scholar] [CrossRef]
  73. Kumar, S.A.; Ananda Kumar, T.D.; Beeraka, N.M.; Pujar, G.V.; Singh, M.; Narayana Akshatha, H.S.; Bhagyalalitha, M. Machine Learning and Deep Learning in Data-Driven Decision Making of Drug Discovery and Challenges in High-Quality Data Acquisition in the Pharmaceutical Industry. Future Med. Chem. 2022, 14, 245–270. [Google Scholar] [CrossRef]
  74. Fu, C.; Chen, Q. The Future of Pharmaceuticals: Artificial Intelligence in Drug Discovery and Development. J. Pharm. Anal. 2025, 15, 101248. [Google Scholar] [CrossRef] [PubMed]
  75. Tang, J.; Aittokallio, T. Network Pharmacology Strategies toward Multi-Target Anticancer Therapies: From Computational Models to Experimental Design Principles. Curr. Pharm. Des. 2014, 20, 23–36. [Google Scholar] [CrossRef]
  76. Qiao, G.; Wang, G.; Li, Y. Causal Enhanced Drug–Target Interaction Prediction Based on Graph Generation and Multi-Source Information Fusion. Bioinformatics 2024, 40, btae570. [Google Scholar] [CrossRef] [PubMed]
  77. Harrold, J.M.; Ramanathan, M.; Mager, D.E. Network-Based Approaches in Drug Discovery and Early Development. Clin. Pharmacol. Ther. 2013, 94, 651. [Google Scholar] [CrossRef]
  78. Duan, P.; Yang, K.; Su, X.; Fan, S.; Dong, X.; Zhang, F.; Li, X.; Xing, X.; Zhu, Q.; Yu, J.; et al. HTINet2: Herb–Target Prediction via Knowledge Graph Embedding and Residual-like Graph Neural Network. Brief. Bioinform. 2024, 25, bbae414. [Google Scholar] [CrossRef]
  79. Yang, F.; Zhang, S.; Pan, W.; Yao, R.; Zhang, W.; Zhang, Y.; Wang, G.; Zhang, Q.; Cheng, Y.; Dong, J.; et al. Signaling Repurposable Drug Combinations against COVID-19 by Developing the Heterogeneous Deep Herb-Graph Method. Brief. Bioinform. 2022, 23, bbac124. [Google Scholar] [CrossRef]
  80. Li, Y.; Qiao, G.; Wang, K.; Wang, G. Drug–Target Interaction Predication via Multi-Channel Graph Neural Networks. Brief. Bioinform. 2022, 23, bbab346. [Google Scholar] [CrossRef] [PubMed]
  81. Ye, S.; Natarajan, M.; Wu, Z.; Gombolay, M.C. Diffusion Models for Multi-Target Adversarial Tracking. In Proceedings of the 2023 International Symposium on Multi-Robot and Multi-Agent Systems (MRS), Boston, MA, USA, 4–5 December 2023; IEEE: Boston, MA, USA, 2023; pp. 142–148. [Google Scholar]
  82. MacLean, F. Knowledge Graphs and Their Applications in Drug Discovery. Expert Opin. Drug Discov. 2021, 16, 1057–1069. [Google Scholar] [CrossRef]
  83. Gong, Z.; Zhang, N.; He, J. KGRN: Knowledge Graph Relational Path Network for Target Prediction of TCM Prescriptions. In Intelligent Computing Theories and Application; Huang, D.-S., Jo, K.-H., Li, J., Gribova, V., Premaratne, P., Eds.; Lecture Notes in Computer Science; Springer International Publishing: Cham, Switzerland, 2021; Volume 12838, pp. 148–161. ISBN 978-3-030-84531-5. [Google Scholar]
  84. Li, H.; Zhou, Y.; Liao, L.; Tan, H.; Li, Y.; Li, Z.; Zhou, B.; Bao, M.; He, B. Pharmacokinetics Effects of Chuanxiong Rhizoma on Warfarin in Pseudo Germ-Free Rats. Front. Pharmacol. 2022, 13, 1022567. [Google Scholar] [CrossRef]
  85. Huang, K.; Chandak, P.; Wang, Q.; Havaldar, S.; Vaid, A.; Leskovec, J.; Nadkarni, G.N.; Glicksberg, B.S.; Gehlenborg, N.; Zitnik, M. A Foundation Model for Clinician-Centered Drug Repurposing. Nat. Med. 2024, 30, 3601–3613. [Google Scholar] [CrossRef]
  86. Ung, C.Y.; Correia, C.; Li, H.; Adams, C.M.; Westendorf, J.J.; Zhu, S. Multiorgan Locked-State Model of Chronic Diseases and Systems Pharmacology Opportunities. Drug Discov. Today 2024, 29, 103825. [Google Scholar] [CrossRef]
  87. Liu, X.; Zhu, F.; Ma, X.H.; Shi, Z.; Yang, S.Y.; Wei, Y.Q.; Chen, Y.Z. Predicting Targeted Polypharmacology for Drug Repositioning and Multi-Target Drug Discovery. Curr. Med. Chem. 2013, 20, 1646–1661. [Google Scholar] [CrossRef]
  88. Zhou, J.-B.; Tang, D.; He, L.; Lin, S.; Lei, J.H.; Sun, H.; Xu, X.; Deng, C.-X. Machine Learning Model for Anti-Cancer Drug Combinations: Analysis, Prediction, and Validation. Pharmacol. Res. 2023, 194, 106830. [Google Scholar] [CrossRef]
  89. Singha, M.; Pu, L.; Srivastava, G.; Ni, X.; Stanfield, B.A.; Uche, I.K.; Rider, P.J.F.; Kousoulas, K.G.; Ramanujam, J.; Brylinski, M. Unlocking the Potential of Kinase Targets in Cancer: Insights from CancerOmicsNet, an AI-Driven Approach to Drug Response Prediction in Cancer. Cancers 2023, 15, 4050. [Google Scholar] [CrossRef] [PubMed]
  90. Wu, Y.; Chen, M.; Qin, Y. Anticancer Drug Response Prediction Integrating Multi-Omics Pathway-Based Difference Features and Multiple Deep Learning Techniques. PLoS Comput. Biol. 2025, 21, e1012905. [Google Scholar] [CrossRef] [PubMed]
  91. Naorem, L.D.; Muthaiyan, M.; Venkatesan, A. Integrated Network Analysis and Machine Learning Approach for the Identification of Key Genes of Triple-negative Breast Cancer. J. Cell. Biochem. 2019, 120, 6154–6167. [Google Scholar] [CrossRef] [PubMed]
  92. Almansour, N.M. Triple-Negative Breast Cancer: A Brief Review About Epidemiology, Risk Factors, Signaling Pathways, Treatment and Role of Artificial Intelligence. Front. Mol. Biosci. 2022, 9, 836417. [Google Scholar] [CrossRef]
  93. Qureshi, R.; Zou, B.; Alam, T.; Wu, J.; Lee, V.H.F.; Yan, H. Computational Methods for the Analysis and Prediction of EGFR-Mutated Lung Cancer Drug Resistance: Recent Advances in Drug Design, Challenges and Future Prospects. IEEE/ACM Trans. Comput. Biol. Bioinf. 2023, 20, 238–255. [Google Scholar] [CrossRef]
  94. Vatansever, S.; Schlessinger, A.; Wacker, D.; Kaniskan, H.Ü.; Jin, J.; Zhou, M.; Zhang, B. Artificial Intelligence and Machine Learning-aided Drug Discovery in Central Nervous System Diseases: State-of-the-arts and Future Directions. Med. Res. Rev. 2021, 41, 1427–1473. [Google Scholar] [CrossRef]
  95. Liu, M.; Srivastava, G.; Ramanujam, J.; Brylinski, M. SynerGNet: A Graph Neural Network Model to Predict Anticancer Drug Synergy. Biomolecules 2024, 14, 253. [Google Scholar] [CrossRef]
  96. Rafiei, F.; Zeraati, H.; Abbasi, K.; Ghasemi, J.B.; Parsaeian, M.; Masoudi-Nejad, A. DeepTraSynergy: Drug Combinations Using Multimodal Deep Learning with Transformers. Bioinformatics 2023, 39, btad438. [Google Scholar] [CrossRef]
  97. Hu, J.; Gao, J.; Fang, X.; Liu, Z.; Wang, F.; Huang, W.; Wu, H.; Zhao, G. DTSyn: A Dual-Transformer-Based Neural Network to Predict Synergistic Drug Combinations. Brief. Bioinform. 2022, 23, bbac302. [Google Scholar] [CrossRef]
  98. Sidorov, P.; Naulaerts, S.; Ariey-Bonnet, J.; Pasquier, E.; Ballester, P.J. Predicting Synergism of Cancer Drug Combinations Using NCI-ALMANAC Data. Front. Chem. 2019, 7, 509. [Google Scholar] [CrossRef] [PubMed]
  99. Preto, A.J.; Matos-Filipe, P.; Mourão, J.; Moreira, I.S. SYNPRED: Prediction of Drug Combination Effects in Cancer Using Different Synergy Metrics and Ensemble Learning. GigaScience 2022, 11, giac087. [Google Scholar] [CrossRef]
  100. Torkamannia, A.; Omidi, Y.; Ferdousi, R. SYNDEEP: A Deep Learning Approach for the Prediction of Cancer Drugs Synergy. Sci. Rep. 2023, 13, 6184. [Google Scholar] [CrossRef]
  101. Baptista, D.; Ferreira, P.G.; Rocha, M. A Systematic Evaluation of Deep Learning Methods for the Prediction of Drug Synergy in Cancer. PLoS Comput. Biol. 2023, 19, e1010200. [Google Scholar] [CrossRef]
  102. El Khili, M.R.; Memon, S.A.; Emad, A. MARSY: A Multitask Deep-Learning Framework for Prediction of Drug Combination Synergy Scores. Bioinformatics 2023, 39, btad177. [Google Scholar] [CrossRef]
  103. Zhao, X.; Xu, J.; Shui, Y.; Xu, M.; Hu, J.; Liu, X.; Che, K.; Wang, J.; Liu, Y. PermuteDDS: A Permutable Feature Fusion Network for Drug-Drug Synergy Prediction. J. Cheminform. 2024, 16, 41. [Google Scholar] [CrossRef] [PubMed]
  104. Gan, Y.; Huang, X.; Guo, W.; Yan, C.; Zou, G. Predicting Synergistic Anticancer Drug Combination Based on Low-Rank Global Attention Mechanism and Bilinear Predictor. Bioinformatics 2023, 39, btad607. [Google Scholar] [CrossRef]
  105. Guo, Y.; Hu, H.; Chen, W.; Yin, H.; Wu, J.; Hsieh, C.-Y.; He, Q.; Cao, J. SynergyX: A Multi-Modality Mutual Attention Network for Interpretable Drug Synergy Prediction. Brief. Bioinform. 2024, 25, bbae015. [Google Scholar] [CrossRef] [PubMed]
  106. Li, X.; Shen, B.; Feng, F.; Li, K.; Tang, Z.; Ma, L.; Li, H. Dual-View Jointly Learning Improves Personalized Drug Synergy Prediction. Bioinformatics 2024, 40, btae604. [Google Scholar] [CrossRef]
  107. Preuer, K.; Lewis, R.P.I.; Hochreiter, S.; Bender, A.; Bulusu, K.C.; Klambauer, G. DeepSynergy: Predicting Anti-Cancer Drug Synergy with Deep Learning. Bioinformatics 2018, 34, 1538–1546. [Google Scholar] [CrossRef] [PubMed]
  108. Zhang, T.; Zhang, L.; Payne, P.R.O.; Li, F. Synergistic Drug Combination Prediction by Integrating Multiomics Data in Deep Learning Models. In Translational Bioinformatics for Therapeutic Development; Markowitz, J., Ed.; Methods in Molecular Biology; Springer: New York, NY, USA, 2021; Volume 2194, pp. 223–238. ISBN 978-1-0716-0848-7. [Google Scholar]
  109. Kuenzi, B.M.; Park, J.; Fong, S.H.; Sanchez, K.S.; Lee, J.; Kreisberg, J.F.; Ma, J.; Ideker, T. Predicting Drug Response and Synergy Using a Deep Learning Model of Human Cancer Cells. Cancer Cell 2020, 38, 672–684.e6. [Google Scholar] [CrossRef]
  110. Wang, J.; Liu, X.; Shen, S.; Deng, L.; Liu, H. DeepDDS: Deep Graph Neural Network with Attention Mechanism to Predict Synergistic Drug Combinations. Brief. Bioinform. 2022, 23, bbab390. [Google Scholar] [CrossRef] [PubMed]
  111. Menden, M.P.; Wang, D.; Mason, M.J.; Szalai, B.; Bulusu, K.C.; Guan, Y.; Yu, T.; Kang, J.; Jeon, M.; Wolfinger, R.; et al. Community Assessment to Advance Computational Prediction of Cancer Drug Combinations in a Pharmacogenomic Screen. Nat. Commun. 2019, 10, 2674. [Google Scholar] [CrossRef]
  112. Zhang, P.; Tu, S. MGAE-DC: Predicting the Synergistic Effects of Drug Combinations through Multi-Channel Graph Autoencoders. PLoS Comput. Biol. 2023, 19, e1010951. [Google Scholar] [CrossRef]
  113. Jiang, P.; Huang, S.; Fu, Z.; Sun, Z.; Lakowski, T.M.; Hu, P. Deep Graph Embedding for Prioritizing Synergistic Anticancer Drug Combinations. Comput. Struct. Biotechnol. J. 2020, 18, 427–438. [Google Scholar] [CrossRef]
  114. Kuru, H.I.; Tastan, O.; Cicek, A.E. MatchMaker: A Deep Learning Framework for Drug Synergy Prediction. IEEE/ACM Trans. Comput. Biol. Bioinf. 2022, 19, 2334–2344. [Google Scholar] [CrossRef]
  115. Yang, J.; Xu, Z.; Wu, W.K.K.; Chu, Q.; Zhang, Q. GraphSynergy: A Network-Inspired Deep Learning Model for Anticancer Drug Combination Prediction. J. Am. Med. Inform. Assoc. 2021, 28, 2336–2345. [Google Scholar] [CrossRef] [PubMed]
  116. Ling, A.; Huang, R.S. Computationally Predicting Clinical Drug Combination Efficacy with Cancer Cell Line Screens and Independent Drug Action. Nat. Commun. 2020, 11, 5848. [Google Scholar] [CrossRef]
  117. Liu, Q.; Xie, L. TranSynergy: Mechanism-Driven Interpretable Deep Neural Network for the Synergistic Prediction and Pathway Deconvolution of Drug Combinations. PLoS Comput. Biol. 2021, 17, e1008653. [Google Scholar] [CrossRef]
  118. Kumar, V.; Banerjee, A.; Roy, K. Machine Learning-Based q-RASAR Approach for the in Silico Identification of Novel Multi-Target Inhibitors against Alzheimer’s Disease. Chemom. Intell. Lab. Syst. 2024, 245, 105049. [Google Scholar] [CrossRef]
  119. El Idrissi, F.; Gressier, B.; Devos, D.; Belarbi, K. A Computational Exploration of the Molecular Network Associated to Neuroinflammation in Alzheimer’s Disease. Front. Pharmacol. 2021, 12, 630003. [Google Scholar] [CrossRef] [PubMed]
  120. Zhao, K.; Xie, H.; Jacobs, T.; Gaggi, N.L.; Fortea, J.; Carlisle, N.B.; Fonzo, G.A.; Pohl, K.M.; Osorio, R.S.; Zhang, Y. Elucidating the Neuropathological and Molecular Heterogeneity of Amyloid-Beta and Tau in Alzheimer’s Disease through Machine Learning and Transcriptomic Integration. bioRxiv 2024. [Google Scholar] [CrossRef]
  121. Parvathy Dharshini, S.A.; Sneha, N.P.; Yesudhas, D.; Kulandaisamy, A.; Rangaswamy, U.; Shanmugam, A.; Taguchi, Y.-H.; Gromiha, M.M. Exploring Plausible Therapeutic Targets for Alzheimer’s Disease usingMulti-Omics Approach, Machine Learning and Docking. Curr. Top. Med. Chem. 2022, 22, 1868–1879. [Google Scholar] [CrossRef] [PubMed]
  122. Nwadiugwu, M.; Onwuekwe, I.; Ezeanolue, E.; Deng, H. Beyond Amyloid: A Machine Learning-Driven Approach Reveals Properties of Potent GSK-3β Inhibitors Targeting Neurofibrillary Tangles. Int. J. Mol. Sci. 2024, 25, 2646. [Google Scholar] [CrossRef] [PubMed]
  123. Yu, S.; Wang, Z.; Nan, J.; Li, A.; Yang, X.; Tang, X. Potential Schizophrenia Disease-Related Genes Prediction Using Metagraph Representations Based on a Protein-Protein Interaction Keyword Network: Framework Development and Validation. JMIR Form. Res. 2023, 7, e50998. [Google Scholar] [CrossRef]
  124. Mohamed, S.K.; Nounu, A.; Nováček, V. Biological Applications of Knowledge Graph Embedding Models. Brief. Bioinform. 2021, 22, 1679–1693. [Google Scholar] [CrossRef]
  125. Nian, Y.; Hu, X.; Zhang, R.; Feng, J.; Du, J.; Li, F.; Bu, L.; Zhang, Y.; Chen, Y.; Tao, C. Mining on Alzheimer’s Diseases Related Knowledge Graph to Identity Potential AD-Related Semantic Triples for Drug Repurposing. BMC Bioinform. 2022, 23, 407. [Google Scholar] [CrossRef]
  126. Wang, H.; Zhang, J.; Lu, Z.; Dai, W.; Ma, C.; Xiang, Y.; Zhang, Y. Identification of Potential Therapeutic Targets and Mechanisms of COVID-19 through Network Analysis and Screening of Chemicals and Herbal Ingredients. Brief. Bioinform. 2022, 23, bbab373. [Google Scholar] [CrossRef]
  127. Li, V.O.K.; Han, Y.; Kaistha, T.; Zhang, Q.; Downey, J.; Gozes, I.; Lam, J.C.K. DeepDrug as an Expert Guided and AI Driven Drug Repurposing Methodology for Selecting the Lead Combination of Drugs for Alzheimer’s Disease. Sci. Rep. 2025, 15, 2093. [Google Scholar] [CrossRef]
  128. Wu, X.; Li, Z.; Chen, G.; Yin, Y.; Chen, C.Y.-C. Hybrid Neural Network Approaches to Predict Drug–Target Binding Affinity for Drug Repurposing: Screening for Potential Leads for Alzheimer’s Disease. Front. Mol. Biosci. 2023, 10, 1227371. [Google Scholar] [CrossRef]
  129. Alhassan, H.H. Integrative Machine Learning and Molecular Simulation Approaches Identify GSK3β Inhibitors for Neurodegenerative Disease Therapy. Sci. Rep. 2025, 15, 21632. [Google Scholar] [CrossRef]
  130. Rodriguez, S.; Hug, C.; Todorov, P.; Moret, N.; Boswell, S.A.; Evans, K.; Zhou, G.; Johnson, N.T.; Hyman, B.T.; Sorger, P.K.; et al. Machine Learning Identifies Candidates for Drug Repurposing in Alzheimer’s Disease. Nat. Commun. 2021, 12, 1033. [Google Scholar] [CrossRef] [PubMed]
  131. Kleandrova, V.V.; Cordeiro, M.N.D.S.; Speck-Planche, A. Perturbation-Theory Machine Learning for Multi-Objective Antibacterial Discovery: Current Status and Future Perspectives. Appl. Sci. 2025, 15, 1166. [Google Scholar] [CrossRef]
  132. Nava Lara, R.A.; Beltrán, J.A.; Brizuela, C.A.; Del Rio, G. Relevant Features of Polypharmacologic Human-Target Antimicrobials Discovered by Machine-Learning Techniques. Pharmaceuticals 2020, 13, 204. [Google Scholar] [CrossRef] [PubMed]
  133. Serafim, M.S.M.; Kronenberger, T.; Oliveira, P.R.; Poso, A.; Honório, K.M.; Mota, B.E.F.; Maltarollo, V.G. The Application of Machine Learning Techniques to Innovative Antibacterial Discovery and Development. Expert Opin. Drug Discov. 2020, 15, 1165–1180. [Google Scholar] [CrossRef]
  134. Speck-Planche, A.; Cordeiro, M.N.D.S. Multi-Target QSAR Approaches for Modeling Protein Inhibitors. Simultaneous Prediction of Activities Against Biomacromolecules Present in Gram-Negative Bacteria. Curr. Top. Med. Chem. 2015, 15, 1801–1813. [Google Scholar] [CrossRef]
  135. Abdel-Basset, M.; Hawash, H.; Elhoseny, M.; Chakrabortty, R.K.; Ryan, M. DeepH-DTA: Deep Learning for Predicting Drug-Target Interactions: A Case Study of COVID-19 Drug Repurposing. IEEE Access 2020, 8, 170433–170451. [Google Scholar] [CrossRef] [PubMed]
  136. Arango-Argoty, G.; Garner, E.; Pruden, A.; Heath, L.S.; Vikesland, P.; Zhang, L. DeepARG: A Deep Learning Approach for Predicting Antibiotic Resistance Genes from Metagenomic Data. Microbiome 2018, 6, 23. [Google Scholar] [CrossRef]
  137. Li, Y.; Xu, Z.; Han, W.; Cao, H.; Umarov, R.; Yan, A.; Fan, M.; Chen, H.; Duarte, C.M.; Li, L.; et al. HMD-ARG: Hierarchical Multi-Task Deep Learning for Annotating Antibiotic Resistance Genes. Microbiome 2021, 9, 40. [Google Scholar] [CrossRef]
  138. Pei, Y.; Shum, M.H.-H.; Liao, Y.; Leung, V.W.; Gong, Y.-N.; Smith, D.K.; Yin, X.; Guan, Y.; Luo, R.; Zhang, T.; et al. ARGNet: Using Deep Neural Networks for Robust Identification and Classification of Antibiotic Resistance Genes from Sequences. Microbiome 2024, 12, 84. [Google Scholar] [CrossRef]
  139. López-Cortés, X.A.; Manríquez-Troncoso, J.M.; Hernández-García, R.; Peralta, D. MSDeepAMR: Antimicrobial Resistance Prediction Based on Deep Neural Networks and Transfer Learning. Front. Microbiol. 2024, 15, 1361795. [Google Scholar] [CrossRef]
  140. Nguyen, M.; Long, S.W.; McDermott, P.F.; Olsen, R.J.; Olson, R.; Stevens, R.L.; Tyson, G.H.; Zhao, S.; Davis, J.J. Using Machine Learning To Predict Antimicrobial MICs and Associated Genomic Features for Nontyphoidal Salmonella. J. Clin. Microbiol. 2019, 57, e01260-18. [Google Scholar] [CrossRef] [PubMed]
  141. Weis, C.; Cuénod, A.; Rieck, B.; Dubuis, O.; Graf, S.; Lang, C.; Oberle, M.; Brackmann, M.; Søgaard, K.K.; Osthoff, M.; et al. Direct Antimicrobial Resistance Prediction from Clinical MALDI-TOF Mass Spectra Using Machine Learning. Nat. Med. 2022, 28, 164–174. [Google Scholar] [CrossRef] [PubMed]
  142. Santos-Júnior, C.D.; Torres, M.D.T.; Duan, Y.; Rodríguez Del Río, Á.; Schmidt, T.S.B.; Chong, H.; Fullam, A.; Kuhn, M.; Zhu, C.; Houseman, A.; et al. Discovery of Antimicrobial Peptides in the Global Microbiome with Machine Learning. Cell 2024, 187, 3761–3778.e16. [Google Scholar] [CrossRef]
  143. Wong, F.; Zheng, E.J.; Valeri, J.A.; Donghia, N.M.; Anahtar, M.N.; Omori, S.; Li, A.; Cubillos-Ruiz, A.; Krishnan, A.; Jin, W.; et al. Discovery of a Structural Class of Antibiotics with Explainable Deep Learning. Nature 2024, 626, 177–185. [Google Scholar] [CrossRef] [PubMed]
  144. Pesesky, M.W.; Hussain, T.; Wallace, M.; Patel, S.; Andleeb, S.; Burnham, C.-A.D.; Dantas, G. Evaluation of Machine Learning and Rules-Based Approaches for Predicting Antimicrobial Resistance Profiles in Gram-Negative Bacilli from Whole Genome Sequence Data. Front. Microbiol. 2016, 7, 1887. [Google Scholar] [CrossRef] [PubMed]
  145. Macesic, N.; Bear Don’t Walk, O.J.; Pe’er, I.; Tatonetti, N.P.; Peleg, A.Y.; Uhlemann, A.-C. Predicting Phenotypic Polymyxin Resistance in Klebsiella Pneumoniae through Machine Learning Analysis of Genomic Data. mSystems 2020, 5, e00656-19. [Google Scholar] [CrossRef]
  146. Stokes, J.M.; Yang, K.; Swanson, K.; Jin, W.; Cubillos-Ruiz, A.; Donghia, N.M.; MacNair, C.R.; French, S.; Carfrae, L.A.; Bloom-Ackermann, Z.; et al. A Deep Learning Approach to Antibiotic Discovery. Cell 2020, 180, 688–702.e13. [Google Scholar] [CrossRef]
  147. Chandrasekaran, S.; Cokol-Cakmak, M.; Sahin, N.; Yilancioglu, K.; Kazan, H.; Collins, J.J.; Cokol, M. Chemogenomics and Orthology-based Design of Antibiotic Combination Therapies. Mol. Syst. Biol. 2016, 12, 872. [Google Scholar] [CrossRef] [PubMed]
  148. Cokol, M.; Li, C.; Chandrasekaran, S. Chemogenomic Model Identifies Synergistic Drug Combinations Robust to the Pathogen Microenvironment. PLoS Comput. Biol. 2018, 14, e1006677. [Google Scholar] [CrossRef] [PubMed]
  149. Lv, J.; Liu, G.; Ju, Y.; Sun, Y.; Guo, W. Prediction of Synergistic Antibiotic Combinations by Graph Learning. Front. Pharmacol. 2022, 13, 849006. [Google Scholar] [CrossRef]
  150. Mason, D.J.; Eastman, R.T.; Lewis, R.P.I.; Stott, I.P.; Guha, R.; Bender, A. Using Machine Learning to Predict Synergistic Antimalarial Compound Combinations with Novel Structures. Front. Pharmacol. 2018, 9, 1096. [Google Scholar] [CrossRef]
  151. Jin, W.; Stokes, J.M.; Eastman, R.T.; Itkin, Z.; Zakharov, A.V.; Collins, J.J.; Jaakkola, T.S.; Barzilay, R. Deep Learning Identifies Synergistic Drug Combinations for Treating COVID-19. Proc. Natl. Acad. Sci. USA 2021, 118, e2105070118. [Google Scholar] [CrossRef]
  152. Olcay, B.; Ozdemir, G.D.; Ozdemir, M.A.; Ercan, U.K.; Guren, O.; Karaman, O. Prediction of the Synergistic Effect of Antimicrobial Peptides and Antimicrobial Agents via Supervised Machine Learning. BMC Biomed. Eng. 2024, 6, 1. [Google Scholar] [CrossRef]
  153. Olayo-Alarcon, R.; Amstalden, M.K.; Zannoni, A.; Bajramovic, M.; Sharma, C.M.; Brochado, A.R.; Rezaei, M.; Müller, C.L. Pre-Trained Molecular Representations Enable Antimicrobial Discovery. Nat. Commun. 2025, 16, 3420. [Google Scholar] [CrossRef]
  154. Zheng, E.J.; Valeri, J.A.; Andrews, I.W.; Krishnan, A.; Bandyopadhyay, P.; Anahtar, M.N.; Herneisen, A.; Schulte, F.; Linnehan, B.; Wong, F.; et al. Discovery of Antibiotics That Selectively Kill Metabolically Dormant Bacteria. Cell Chem. Biol. 2024, 31, 712–728.e9. [Google Scholar] [CrossRef]
  155. Rannon, E.; Shaashua, S.; Burstein, D. DRAMMA: A Multifaceted Machine Learning Approach for Novel Antimicrobial Resistance Gene Detection in Metagenomic Data. Microbiome 2025, 13, 67. [Google Scholar] [CrossRef] [PubMed]
  156. Wang, T.; Pulkkinen, O.I.; Aittokallio, T. Target-Specific Compound Selectivity for Multi-Target Drug Discovery and Repurposing. Front. Pharmacol. 2022, 13, 1003480. [Google Scholar] [CrossRef]
  157. Mukherjee, A.; Abraham, S.; Singh, A.; Balaji, S.; Mukunthan, K.S. From Data to Cure: A Comprehensive Exploration of Multi-Omics Data Analysis for Targeted Therapies. Mol. Biotechnol. 2025, 67, 1269–1289. [Google Scholar] [CrossRef]
  158. Lim, H.; He, D.; Qiu, Y.; Krawczuk, P.; Sun, X.; Xie, L. Rational Discovery of Dual-Indication Multi-Target PDE/Kinase Inhibitor for Precision Anti-Cancer Therapy Using Structural Systems Pharmacology. PLoS Comput. Biol. 2019, 15, e1006619. [Google Scholar] [CrossRef] [PubMed]
  159. Liu, X.; Ye, K.; Van Vlijmen, H.W.T.; Emmerich, M.T.M.; IJzerman, A.P.; Van Westen, G.J.P. DrugEx v2: De Novo Design of Drug Molecules by Pareto-Based Multi-Objective Reinforcement Learning in Polypharmacology. J. Cheminform. 2021, 13, 85. [Google Scholar] [CrossRef] [PubMed]
  160. Yang, Y.; Chen, G.; Li, J.; Li, J.; Zhang, O.; Zhang, X.; Li, L.; Hao, J.; Wang, E.; Heng, P.-A. Enabling Target-Aware Molecule Generation to Follow Multi Objectives with Pareto MCTS. Commun. Biol. 2024, 7, 1074. [Google Scholar] [CrossRef]
  161. Yao, L.; Jin, Z.; Mao, C.; Zhang, Y.; Luo, Y. Traditional Chinese Medicine Clinical Records Classification with BERT and Domain Specific Corpora. J. Am. Med. Inform. Assoc. 2019, 26, 1632–1636. [Google Scholar] [CrossRef] [PubMed]
  162. Liu, L.-C.; Ho, M.-Y.; Su, B.-H.; Wang, S.-Y.; Hsu, M.-T.; Tseng, Y.J. PanGPCR: Predictions for Multiple Targets, Repurposing and Side Effects. Bioinformatics 2021, 37, 1184–1186. [Google Scholar] [CrossRef]
  163. Zięba, A.; Stępnicki, P.; Matosiuk, D.; Kaczor, A.A. What Are the Challenges with Multi-Targeted Drug Design for Complex Diseases? Expert Opin. Drug Discov. 2022, 17, 673–683. [Google Scholar] [CrossRef]
  164. Ravikumar, B.; Aittokallio, T. Improving the Efficacy-Safety Balance of Polypharmacology in Multi-Target Drug Discovery. Expert Opin. Drug Discov. 2018, 13, 179–192. [Google Scholar] [CrossRef]
  165. Ramsay, R.R.; Popovic-Nikolic, M.R.; Nikolic, K.; Uliassi, E.; Bolognesi, M.L. A Perspective on Multi-target Drug Discovery and Design for Complex Diseases. Clin. Transl. Med. 2018, 7, e3. [Google Scholar] [CrossRef]
  166. Rodríguez-Pérez, R.; Bajorath, J. Interpretation of Machine Learning Models Using Shapley Values: Application to Compound Potency and Multi-Target Activity Predictions. J. Comput. Aided Mol. Des. 2020, 34, 1013–1026. [Google Scholar] [CrossRef]
  167. Simões, R.S.; Maltarollo, V.G.; Oliveira, P.R.; Honorio, K.M. Transfer and Multi-Task Learning in QSAR Modeling: Advances and Challenges. Front. Pharmacol. 2018, 9, 74. [Google Scholar] [CrossRef] [PubMed]
  168. Qiang, B.; Lai, J.; Jin, H.; Zhang, L.; Liu, Z. Target Prediction Model for Natural Products Using Transfer Learning. Int. J. Mol. Sci. 2021, 22, 4632. [Google Scholar] [CrossRef] [PubMed]
  169. Bettanti, A.; Beccari, A.R.; Biccarino, M. Exploring the Future of Biopharmaceutical Drug Discovery: Can Advanced AI Platforms Overcome Current Challenges? Discov. Artif. Intell. 2024, 4, 102. [Google Scholar] [CrossRef]
Figure 1. Organizational Framework of the Review. This mind map provides a comprehensive overview of the article’s structure, illustrating the logical flow from the introduction of multi-target drug discovery to the fundamental concepts, advanced machine learning techniques, real-world applications, and concluding with challenges and future perspectives.
Figure 1. Organizational Framework of the Review. This mind map provides a comprehensive overview of the article’s structure, illustrating the logical flow from the introduction of multi-target drug discovery to the fundamental concepts, advanced machine learning techniques, real-world applications, and concluding with challenges and future perspectives.
Pharmaceutics 17 01186 g001
Figure 2. Comparison of Therapeutic Strategies. (A) The classic “one-drug, one-target” model, where a single drug is designed to modulate a specific target to achieve a therapeutic effect. Off-target binding can lead to unintended side effects. (B) A systems biology view, which considers how a drug’s interaction with a single target can propagate through a complex biological network, leading to both desired therapeutic effects and potential side effects. (C) The systems pharmacology approach, where a single drug is designed to act on multiple targets or a network of targets to produce a synergistic therapeutic effect and restore network stability, thus minimizing side effects.
Figure 2. Comparison of Therapeutic Strategies. (A) The classic “one-drug, one-target” model, where a single drug is designed to modulate a specific target to achieve a therapeutic effect. Off-target binding can lead to unintended side effects. (B) A systems biology view, which considers how a drug’s interaction with a single target can propagate through a complex biological network, leading to both desired therapeutic effects and potential side effects. (C) The systems pharmacology approach, where a single drug is designed to act on multiple targets or a network of targets to produce a synergistic therapeutic effect and restore network stability, thus minimizing side effects.
Pharmaceutics 17 01186 g002
Figure 3. Representative Machine Learning Pipelines for Drug-Target Prediction. The figure illustrates three different computational workflows. (A) A classical ML pipeline that relies on feature engineering, where chemical structures and protein sequences are converted into handcrafted features (e.g., FP2 fingerprints, PsePSSM) before being used to train traditional models like RF, Lasso, or SVM for prediction. (B) A DL pipeline that utilizes GNNs for representation learning, where drugs and proteins are represented as nodes in a graph and GNNs, including variants like graph convolutional networks (GCNs) and graph attention networks (GATs), automatically learn features to predict DTI. (C) A modern DL pipeline that integrates advanced architectures, such as a residual GNN for compound representation and an attention module for protein sequence embedding, which are then fused and processed by a multi-layer perceptron (MLP) for final prediction. This approach leverages the strengths of both graph-based and attention-based models to capture complex relationships.
Figure 3. Representative Machine Learning Pipelines for Drug-Target Prediction. The figure illustrates three different computational workflows. (A) A classical ML pipeline that relies on feature engineering, where chemical structures and protein sequences are converted into handcrafted features (e.g., FP2 fingerprints, PsePSSM) before being used to train traditional models like RF, Lasso, or SVM for prediction. (B) A DL pipeline that utilizes GNNs for representation learning, where drugs and proteins are represented as nodes in a graph and GNNs, including variants like graph convolutional networks (GCNs) and graph attention networks (GATs), automatically learn features to predict DTI. (C) A modern DL pipeline that integrates advanced architectures, such as a residual GNN for compound representation and an attention module for protein sequence embedding, which are then fused and processed by a multi-layer perceptron (MLP) for final prediction. This approach leverages the strengths of both graph-based and attention-based models to capture complex relationships.
Pharmaceutics 17 01186 g003
Figure 4. A Comparative Overview of Machine Learning in Multi-Target Drug Discovery. This figure summarizes the key trends and strategies of ML applications across four critical therapeutic areas.
Figure 4. A Comparative Overview of Machine Learning in Multi-Target Drug Discovery. This figure summarizes the key trends and strategies of ML applications across four critical therapeutic areas.
Pharmaceutics 17 01186 g004
Table 1. Representative Data Sources for Machine Learning in Multi-Target Drug Discovery.
Table 1. Representative Data Sources for Machine Learning in Multi-Target Drug Discovery.
Database NameData TypeBrief DescriptionURL/Reference
TTDTherapeutic targets, drugs, diseasesProvides comprehensive information on known and explored therapeutic protein and nucleic acid targets, their associated diseases, pathways, and corresponding drugs.https://idrblab.org/ttd/ (accessed on 8 August 2025)
KEGGGenomics, pathways, diseases, drugsA knowledge base that links genomic information with higher-level functional information, such as biological pathways, diseases, and drug networks.https://www.genome.jp/kegg/ (accessed on 8 August 2025)
PDBProtein and nucleic acid 3D structuresA global archive for the experimentally determined 3D structures of biological macromolecules, including proteins and nucleic acids.https://www.rcsb.org/ (accessed on 8 August 2025)
DrugBankDrug-target, chemical, pharmacological dataA comprehensive resource that combines detailed drug data with extensive information on drug targets, mechanisms of action, and pathways.https://go.drugbank.com/ (accessed on 8 August 2025)
ChEMBLBioactivity, chemical, genomic dataA manually curated database of bioactive drug-like small molecules, their bioactivities, and associated targets, extracted from medicinal chemistry literature.https://www.ebi.ac.uk/chembl/ (accessed on 8 August 2025)
BindingDBProtein-ligand binding affinitiesA public database of experimentally measured binding affinities, focusing on interactions between small molecules and proteins.http://www.bindingdb.org/ (accessed on 8 August 2025)
STITCHChemical-protein interactionsA database of known and predicted interactions between chemicals and proteins, integrating data from curated sources, text mining, and experiments.http://stitch.embl.de/ (accessed on 8 August 2025)
PubChemChemical information, bioactivityA massive repository of chemical compounds and their biological activities, including structures, bioassay results, and patents.https://pubchem.ncbi.nlm.nih.gov/ (accessed on 8 August 2025)
STRINGProtein–protein interactions (PPI)A database of known and predicted PPIs. It includes both physical and functional associations derived from experiments and genomic context.https://string-db.org/ (accessed on 8 August 2025)
PharmGKBPharmacogenomic dataA comprehensive knowledge base that curates information on how genetic variations influence an individual’s response to drugs, linking genes, drugs, and diseases.https://www.pharmgkb.org/ (accessed on 8 August 2025)
ClinicalTrials.govClinical trial informationA public database maintained by the U.S. National Institutes of Health, containing a registry and results of clinical studies from around the world.https://clinicaltrials.gov/ (accessed on 8 August 2025)
DisGeNETGene-disease associationsA discovery platform that integrates information on human gene-disease associations from various data sources and expert curations.https://www.disgenet.org/ (accessed on 8 August 2025)
ReactomeBiological pathwaysAn open-source, curated database of biological pathways and molecular processes in humans, providing detailed information on molecular interactions and events.https://reactome.org/ (accessed on 8 August 2025)
DrugCombDBDrug combination dataA specialized database for drug combinations, featuring data on synergy, antagonism, and additivity to guide drug combination research.https://drugcomb.org/ (accessed on 8 August 2025)
NCI-ALMANACDrug combination screening dataA large-scale cancer cell line drug combination screening dataset maintained by the National Cancer Institute (NCI).https://dtp.cancer.gov/ncialmanac/initializePage.do (accessed on 8 August 2025)
O’Neil (Dataset)Drug combination screening dataA key dataset for drug combination synergy prediction, published by O’Neil et al. in a 2016 study.[45]
Table 2. Representative Studies on Machine Learning Applications for Drug Combination and Synergy Prediction in Oncology.
Table 2. Representative Studies on Machine Learning Applications for Drug Combination and Synergy Prediction in Oncology.
Data SourcesML ApproachesMain Research FocusKey FindingsRef.
AZ-DREAM Challenges synergy data and an independent validation set from DrugCombDBA GNN model named SynerGNet, with data augmentationTo develop a GNN model to accurately predict the synergistic effects of drug pairs against cancer cell lines.SynerGNet achieved a balanced accuracy of 0.68, outperforming traditional ML. Data augmentation further improved accuracy to 0.73, and the model showed strong performance on unseen data from DrugCombDB.[95]
DrugCombDB and Oncology-Screen datasetsDeepTraSynergy, a multimodal DL approach using transformers and a multitask learning frameworkTo predict drug combination synergy using a new DL model that integrates multimodal data, including drug-target, protein-protein, and cell-target interactions.DeepTraSynergy outperformed classical and state-of-the-art models, achieving accuracies of 0.7715 and 0.8052 on the DrugCombDB and Oncology-Screen datasets, respectively. The study confirmed that integrating PPI networks significantly improved prediction.[96]
DDS data and five independent datasetsDTSyn, a deep neural network (DNN) model using a multi-head attention mechanismTo identify novel drug combinations and understand the mechanisms of drug synergy from a chemical–gene–tissue interaction perspective.DTSyn achieved the highest area under the curve (AUC) for the receiver operating characteristic curve (ROC) and best true positive rate (TPR) compared to competing methods. The ablation study confirmed the contribution of both transformer encoder blocks, and the model showed improved interpretability by extracting interactions among chemicals and cancer cell lines.[97]
NCI-ALMANAC datasetRF, XGBoostTo predict the synergy of unseen cancer drug combinations using a large-scale modeling study to reduce the need for in vitro tests.The models predicted synergy with high accuracy. The study also found that certain drug classes—alkylating agents, tyrosine kinase inhibitors, and topoisomerase inhibitors—were better predicted, and that restricting predictions to the most reliable ones significantly decreased the error rate.[98]
NCI-ALMANAC datasetSYNPRED, an interdisciplinary approach leveraging specifically designed ensembles of AI algorithmsTo develop a robust ensemble learning model for predicting anticancer drug synergy while also focusing on data interpretability. The study also aimed to determine the most appropriate synergy metric.SYNPRED achieved state-of-the-art performance in both classification and regression models, particularly when using the Combination Sensitivity Score. The study also provided insights into which synergy metrics are most effective and achieved data interpretability using feature importance approaches.[99]
NCI-ALMANAC datasetSYNDEEP, a DNN-based binary classification modelTo develop a novel DNN model for predicting synergistic drug combinations in cancer therapy.The proposed DNN model achieved high accuracy (92.21%) and AUC (97.32%) in tenfold cross-validation. The integration of different types of physicochemical and genomic features was found to lead to more accurate predictions.[100]
NCI-ALMANAC datasetDL methods benchmarkingSystematically evaluate the impact of various methodological choices of multimodal DL models for predicting drug synergy in cancer.Feature selection based on biological knowledge improved performance. Drug features were more predictive than a baseline model. Molecular fingerprints performed slightly better than learned representations. DL outperformed other ML methods, and an ensemble combining top DL and ML models provided further performance improvements. The models can also learn biologically meaningful associations between drug and cell line features.[101]
CellMiner database and DrugComb datasetsMARSY, a multitask DL model using two encoders to capture the interplay between drug pairs and cell linesTo develop a computational model to accurately predict cancer drug-pair synergy scores by imputing missing values in sparse datasets.MARSY outperformed state-of-the-art and traditional ML models. It successfully predicted synergy scores for new cancer drug-pair cell line combinations, and these novel predictions were validated using independent studies.[102]
O’Neil and NCI-ALMANAC datasets; PubChem and GDSC databasesPermuteDDS, a Permutable feature fusion networkTo develop an effective computational method for predicting drug–drug synergy (DDS) in cancer.PermuteDDS exhibited superior performance on two benchmark datasets and showed good generalization performance on an independent test set. The permutable fusion mechanism was found to be an effective way to combine drug and cell line features.[103]
DrugComb, Chembl, HURI datasets and othersDGSSynADR, a DL method using a low-rank global attention (LRGA) model and a bilinear predictorTo develop a new DL method to predict synergistic anticancer drug combinations, focusing on improving model generalization and interpretability.DGSSynADR achieved better performance compared to seven competitive methods. The LRGA model and bilinear predictor were found to be key to improving prediction performance, and a Smooth L1 loss function helped avoid gradient explosion.[104]
DrugComb, O’Neil, ALMANAC, Oncology Screen, DrugCombDB datasets and othersSynergyX, a multi-modality mutual attention network with a convolution-augmented attention structureTo develop a computational method for interpretable anti-tumor drug synergy prediction by modeling complex biological and drug interactions.SynergyX showed superior predictive accuracy compared to other models. It also demonstrated multidimensional interpretability, identifying promising drug combinations for potential lung cancer treatment and uncovering drug-gene interactions.[105]
DrugComb and CCLE databasesJointSyn, a dual-view jointly learning modelTo develop a model for personalized drug synergy prediction that is accurate and robust, especially for cross-dataset predictions.JointSyn outperformed existing models in predictive accuracy and robustness. The dual-view approach captured complementary synergy-related characteristics. The model’s generalization ability was improved with fine-tuning, and it was used to generate a drug synergy atlas for pan-cancer.[106]
A large-scale oncology screen published by Merck & Co.DeepSynergy, a DL model using conical layersTo apply DL to predict anticancer drug synergy, using chemical and genomic information.DeepSynergy significantly outperformed other ML methods, with a 7.2% improvement over the next best method. It achieved a Pearson correlation of 0.73 and a high AUC of 0.90 for classifying novel combinations.[107]
Multiomics datasets from TCGA and othersAuDNNsynergy, a DL model that uses three autoencoders to encode omics dataTo develop a novel DL model for predicting the synergy of pairwise drug combinations in cancer by integrating multiomics data.AuDNNsynergy outperformed four state-of-the-art approaches, specifically in terms of rank correlation metrics.[108]
CCLE, GDSC datasets and othersDrugCell, an DL model that integrates tumor genotypes and drug structure to predict responseTo develop an interpretable DL model for predicting drug response and designing synergistic drug combinations in cancer.DrugCell accurately predicts drug responses, stratifies clinical outcomes, and successfully identifies synergistic drug combinations, which were validated through in vitro and in vivo experiments.[109]
DrugBank, CCLE databases and an independent test set released by AstraZenecaDeepDDS, a DL model based on a GNN with an attention mechanismTo develop a GNN-based DL model to identify synergistic drug combinations for specific cancer cells.DeepDDS achieved better performance than both classical and other DL methods on a benchmark dataset. It also outperformed competitive methods by more than 16% in predictive precision on an independent test set. The model’s graph attention network provided interpretability by revealing important chemical substructures.[110]
AstraZeneca’s drug combination datasetDREAM challenge, a community-based, competitive benchmarking frameworkTo evaluate computational strategies for predicting synergistic drug pairs and biomarkers using a large-scale pharmacogenomic screen.Winning methods achieved an accuracy matching biological replicates for over 60% of combinations. The study also identified a genomic rationale for some synergy predictions, but noted that 20% of combinations were poorly predicted by all methods.[111]
O’Neil, ALMANAC, CLOUD and FORCINA datasetsMGAE-DC, a multi-channel graph autoencoder (MGAE) modelTo develop a MGAE model that can predict the synergistic effects of drug combinations by leveraging not only synergistic data but also additive and antagonistic data.MGAE-DC consistently outperformed state-of-the-art methods on four benchmark datasets. The model’s approach of integrating non-synergistic data and using an attention mechanism to capture invariant patterns improved its generalization performance and predictive accuracy.[112]
O’Neil dataset; various online databases and published literatureA GCN model to solve a link prediction taskTo develop a GCN model to predict cell line-specific synergistic anticancer drug combinations by integrating multiple biological networks.The GCN model accurately predicted synergistic drug combinations, with a mean AUC of 0.84 across 39 cell lines. An in-depth literature survey validated many of the top predicted combinations, confirming their synergistic anti-tumor activity.[113]
DrugComb dataset; cell-line omics databaseMatchMaker, a DL framework that uses drug chemical structure and gene expression profilesTo develop a DL model to predict drug synergy scores with high accuracy by using the largest available dataset.MatchMaker showed significant improvements over state-of-the-art models, with up to ~15% better correlation and ~33% lower mean squared error (MSE). The study also identified drug pairs and cell types that were harder to predict and presented novel candidate pairs.[114]
DrugCombDB and Oncology-Screen datasetsGraphSynergy, a DL framework using a spatial-based GCN and an attention componentTo develop an end-to-end DL framework for predicting synergistic anticancer drug combinations by encoding high-order topological relationships in a PPI network.GraphSynergy outperformed classic and state-of-the-art models on two datasets, with accuracy values of 0.7553 and 0.7557. The study also found that the model’s attention mechanism highlighted pivotal proteins with relevant molecular functions, improving interpretability.[115]
Monotherapy data from high-throughput cancer cell line screens, and efficacy data from over 5000 in vitro drug combinations and 26 clinical trialsIDACombo, a method based on the principle of independent drug action (IDA)To develop a method to predict the clinical efficacy of cancer drug combinations using monotherapy cell line screen data.IDACombo’s predictions showed high agreement with measured efficacies both in vitro (Pearson’s correlation = 0.93) and in clinical trials (accuracy > 84%). The method provides a framework for prioritizing new combinations.[116]
Drug combination synergy score dataset; DrugBank and ChEMBL datasets; CCLE and GDSC databasesTranSynergy, a knowledge-enabled DL model using a self-attention transformerTo develop a mechanism-driven, interpretable DNN for synergistic drug combination prediction and pathway deconvolution, with a focus on cancer treatments.TranSynergy outperformed the state-of-the-art method in performance and interpretability. It revealed novel pathways associated with synergistic combinations and predicted new high-confidence synergistic combinations for ovarian cancer.[117]
Note: The table provides an overview of key studies, including their data sources, machine learning (ML) approaches, main research focus, and key findings.
Table 3. Representative Studies on Machine Learning Applications for Multi-Target Drug Discovery in Neurodegenerative Diseases.
Table 3. Representative Studies on Machine Learning Applications for Multi-Target Drug Discovery in Neurodegenerative Diseases.
Data SourcesML ApproachesMain Research FocusKey FindingsRef.
DrugBank, DrugCentral, ChEMBL, BindingDB datasets and othersDeepDrug, a GNN modelTo propose a novel AI-driven drug repurposing methodology to identify an effective combination of approved drugs for AD.DeepDrug successfully identified a five-drug lead combination that targets multiple AD-related pathologies, demonstrating a novel expert-guided, AI-driven method for drug combination discovery in neurodegenerative diseases.[127]
STRING and DrugBank databasesHNNDTA, a hybrid neural network for drug-target affinity (DTA) predictionTo propose a hybrid neural network for DTA prediction to facilitate drug repurposing for AD by identifying potential leads targeting the sigma-1 receptor.The study identified haloperidol and bromperidol as lead compounds for AD treatment, proposing a new computer-aided drug design approach that is faster and more economical and has the potential for multi-target action.[128]
BindingDB, ChEMBL, PubChem, and MPD3 datasetsML virtual screening + molecular dynamicsTo identify potential GSK3β inhibitors for treating neurodegenerative diseases, such as AD and PD, using a combined ML and molecular simulation approach.The study identified three compounds with strong binding scores, with ZINC136900288 showing the highest affinity. These compounds feature novel chemical scaffolds and may serve as promising candidates for future experimental validation as GSK3β inhibitors.[129]
ChEMBL database; Synapse portal dataset and othersDRIAD, a ML framework that links gene expression to AD pathologyTo develop a ML framework for identifying drug repurposing candidates for AD.The DRIAD method produced a ranked list of potential drug candidates for AD, providing a systematic way to nominate drugs for future clinical trials.[130]
Note: The table structure and description are consistent with those of Table 2.
Table 4. Representative Studies on Machine Learning Applications for Combating Infectious Diseases and Antimicrobial Resistance.
Table 4. Representative Studies on Machine Learning Applications for Combating Infectious Diseases and Antimicrobial Resistance.
Data SourcesML ApproachesMain Research FocusKey FindingsRef.
CARD, ARDB, and UNIPROT databesesTwo DL models, DeepARG-SS and DeepARG-LS, were developed to predict antibiotic resistance genes (ARG)To propose a DL approach for accurately predicting antibiotic resistance genes from metagenomic data to address the limitations of traditional bioinformatics methods.The DeepARG models demonstrated high precision and recall in predicting ARG and consistently outperformed the typical “best hit” approach by yielding lower false negative rates.[136]
The HMD-ARG-DB, a curated multi-label ARG databaseHMD-ARG, an end-to-end Hierarchical Multi-task DL framework for ARG annotationTo develop a multi-task DL framework for the accurate and detailed annotation of ARG from sequence data.The HMD-ARG method demonstrates superior performance over state-of-the-art methods and can simultaneously identify an ARG’s class, resistance mechanism, and gene mobility.[137]
ARGNet-DB, a dataset containing 48,615 amino acid sequencesARGNet, a DNN that combines an unsupervised learning autoencoder with a CNN for classificationTo develop a robust, alignment-free DNN to identify and classify a broad range of ARG from sequence data.ARGNet demonstrated superior performance over other DL models. The model is flexible, efficient, and accurate, making it suitable for both targeted and metagenomic sequencing data.[138]
DRIAMS databaseMSDeepAMR, a DNN modelTo propose a DNN model to predict AMR from raw mass spectrometry data.The MSDeepAMR models showed good classification performance, with AUROC values above 0.83 in most cases. Additionally, adapted models improved AUROC by up to 20% compared to models trained on external data alone.[139]
NARMS program for genomes and metadataXGBoost-based models were used to predict minimum inhibitory concentration (MIC) from whole-genome sequencesTo develop highly accurate ML models for predicting antimicrobial MIC and identifying associated genomic features in nontyphoidal Salmonella.The models achieved an average accuracy of 95% for predicting MIC. The study also demonstrated that highly accurate models could be generated with a small number of genomes and that the approach is stable over time.[140]
DRIAMS databaseCalibrated classifiers were trained to predict AMR from MALDI-TOF mass spectra profilesTo develop a ML approach to accelerate the determination of AMR directly from clinical MALDI-TOF mass spectra.The models showed strong predictive potential with AUROC values of 0.80, 0.74, and 0.74 for S. aureus, E. coli, and K. pneumoniae, respectively. A clinical case study showed that this approach would have been beneficial in a majority of cases by providing a faster and more accurate treatment recommendation.[141]
Microbiome datasetsA ML approach that uses RF to predict and catalog antimicrobial peptides (AMPs)To discover and catalog novel AMPs from the global microbiome to address the antibiotic-resistance crisis.The research resulted in the AMPSphere catalog of nearly one million non-redundant peptides. Validation showed that 79 out of 100 tested peptides were active against clinically relevant drug-resistant pathogens.[142]
Chemical + genomic datasetsExplainable DL using ensembles of GNN and explainable graph algorithmsTo use an explainable DL approach to discover a novel structural class of antibiotics to combat antibiotic resistance.The method successfully identified a new structural class of antibiotics that are selective against drug-resistant pathogens like MRSA and VRE and are effective in mouse models of infection.[143]
Whole-genome sequence (WGS) data from Enterobacteriaceae isolatesA ML algorithm was compared against a rules-based approach for predicting resistance profilesTo evaluate and compare the performance of rules-based and ML approaches for predicting AMR from WGS data.Both the rules-based and ML approaches achieved high agreement (89.0% and 90.3%, respectively) with standard phenotypic diagnostics. The study also identified specific sources of disagreement for each method.[144]
Bacterial WGS datasets and phenotypes dataSeveral ML algorithms, including logistic regression, RF, SVC, and GBTCTo apply ML to WGS data to predict phenotypic polymyxin resistance in Klebsiella pneumoniae CG258.The ML approach with a reference-based genomic data representation outperformed the rules-based approach. The models also correctly identified known resistance genes and suggested potential novel determinants.[145]
Chemical libraries: Drug Repurposing Hub and ZINC15 databaseA DNN model was trained to predict molecules with antibacterial activityTo use DL model to discover new, structurally divergent antibiotics to combat antibiotic resistance.The DL model successfully identified a molecule, halicin, which is effective against multi-drug resistant pathogens. The approach also identified eight other new antibacterial compounds from a large chemical library.[146]
Chemogenomics dataINDIGO, a computational approach that predicts synergistic or antagonistic interactionsTo develop a computational method to predict effective antibiotic combination therapies that counter drug resistance.The INDIGO approach significantly outperformed existing methods in predicting antibiotic interactions in E. coli and was successfully used to estimate drug-interaction outcomes in other pathogens like M. tuberculosis and S. aureus.[147]
Chemogenomic profiles of drugs and metabolic perturbationsMAGENTA, a computational framework that predicts drug–drug interactionsTo develop a computational framework to identify robust synergistic antibiotic combinations by predicting their efficacy in diverse microenvironments.The framework successfully identified synergistic antibiotic combinations that are effective across different environments and accurately predicted changes in drug efficacy.[148]
Antibiotic combination and PPI network dataA graph learning framework that combines network proximity with network propagation and a graph regularization modelTo develop a graph learning framework to predict synergistic antibiotic combinations.The model showed better performance and interpretability compared to existing methods for predicting synergistic antibiotic combinations.[149]
Antimalarial compound libraries + in vitro assaysCoSynE: a ML model for predicting antimalarial combination synergyTo use ML to predict novel, synergistic antimalarial drug combinations with a focus on addressing drug resistance.CoSynE successfully predicted synergistic combinations with significant enrichment over random selection, including with entirely novel compounds.[150]
DTI and antiviral data, drug–drug combination datasetsA neural network architecture that jointly learns DTI and DDSTo use DL to identify synergistic drug combinations for COVID-19 with limited combination data.The model performed significantly better than previous methods and successfully predicted two synergistic drug combinations that were validated in vitro.[151]
AMP and antimicrobial agent synergy datasetsSupervised ML classifiers (RF, SVM) for AMP synergyTo predict the synergistic effects of combining AMP and antimicrobial agents using ML to reduce experimental effort and cost.The models achieved a high accuracy of 76.92% in predicting synergistic effects. The analysis also revealed the most important features for prediction, which include the target microbial species and the MIC of the agents.[152]
Large-scale chemical screening databasesMolE, a self-supervised DL framework to learn task-independent molecular representationsTo develop a lightweight computational strategy for antimicrobial discovery using pre-trained molecular representations.The model successfully identified and experimentally confirmed three human-targeted drugs as growth inhibitors of Staphylococcus aureus that were structurally distinct from existing antibiotics.[153]
Broad Institute and other compound databasesDL models were used for virtual screens and toxicity filteringTo discover non-toxic antibiotics that are effective against metabolically dormant bacteria, which are associated with chronic infections and resistance.The approach successfully identified semapimod, an anti-inflammatory drug that selectively kills stationary-phase bacteria by disrupting the outer membrane.[154]
Global-scale genomic and metagenomic samplesDRAMMA, a ML method that uses protein properties, genomic context, and evolutionary patterns to detect novel genesTo develop a ML method for detecting novel ARGs in metagenomic data.DRAMMA demonstrated robust predictive performance and holds promise for the discovery of novel ARGs that lack sequence similarity to any known resistance genes.[155]
Note: The table structure and description are consistent with those of Table 2.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bi, X.; Wang, Y.; Wang, J.; Liu, C. Machine Learning for Multi-Target Drug Discovery: Challenges and Opportunities in Systems Pharmacology. Pharmaceutics 2025, 17, 1186. https://doi.org/10.3390/pharmaceutics17091186

AMA Style

Bi X, Wang Y, Wang J, Liu C. Machine Learning for Multi-Target Drug Discovery: Challenges and Opportunities in Systems Pharmacology. Pharmaceutics. 2025; 17(9):1186. https://doi.org/10.3390/pharmaceutics17091186

Chicago/Turabian Style

Bi, Xueyuan, Yangyang Wang, Jihan Wang, and Cuicui Liu. 2025. "Machine Learning for Multi-Target Drug Discovery: Challenges and Opportunities in Systems Pharmacology" Pharmaceutics 17, no. 9: 1186. https://doi.org/10.3390/pharmaceutics17091186

APA Style

Bi, X., Wang, Y., Wang, J., & Liu, C. (2025). Machine Learning for Multi-Target Drug Discovery: Challenges and Opportunities in Systems Pharmacology. Pharmaceutics, 17(9), 1186. https://doi.org/10.3390/pharmaceutics17091186

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop