View Article

Abstract

Drug–drug interactions (DDIs) pose a significant clinical challenge, contributing to adverse drug events (ADEs), hospitalizations, and increased healthcare costs. Conventional DDI detection methods such as in vitro assays, in vivo studies, and post-marketing surveillance—provide valuable mechanistic insights but are limited by high cost, time requirements, and incomplete coverage of possible drug combinations. The rapid expansion of available pharmaceuticals has intensified the need for scalable, accurate, and timely predictive approaches. Artificial intelligence (AI) and machine learning (ML) have emerged as transformative tools capable of integrating large, heterogeneous datasets spanning chemical structures, pharmacokinetics, pharmacodynamics, electronic health records (EHRs), and biomedical literature to uncover complex interaction patterns that may not be evident through traditional methods. This review provides a comprehensive overview of AI-based predictive modeling techniques for DDI identification, encompassing traditional ML algorithms, deep learning architectures, graph-based models, and natural language processing (NLP) approaches. It examines key data sources, including structured databases (e.g., DrugBank, ChEMBL, KEGG), real-world clinical data, and literature mining, and discusses strategies for data preprocessing, feature engineering, and model evaluation. Special attention is given to challenges such as data sparsity, class imbalance, lack of interpretability, and limited generalizability to novel drug combinations. Emerging solutions—including explainable AI (XAI), transfer learning, multimodal data integration, and federated learning—are highlighted as promising directions to enhance transparency, robustness, and clinical applicability. Applications of AI-driven DDI prediction span clinical decision support systems, drug development pipelines, and pharmacovigilance frameworks, with case studies demonstrating reductions in medication errors and improvements in patient safety. The review also addresses regulatory perspectives, ethical considerations, and integration challenges in clinical workflows. Ultimately, AI-based predictive modeling offers a powerful, adaptive, and scalable approach to mitigating DDI risks, supporting precision medicine, and advancing drug safety across healthcare systems.

Keywords

Drug drug interactions, Artificial intelligence, Machine learning, Deep learning, Predictive modeling , Pharmacovigilance, Explainable AI

Introduction

Drug-drug interactions (DDIs) occur when the pharmacological effect or pharmacokinetic profile of one drug is altered by the presence of another, potentially resulting in diminished efficacy or increased toxicity (1). These interactions can be pharmacokinetic, involving changes in drug absorption, distribution, metabolism, or excretion, or pharmacodynamic, where drugs influence each other's effects at the receptor or cellular level (2). DDIs represent a significant challenge in clinical practice, contributing substantially to adverse drug reactions, hospitalizations, and increased healthcare costs worldwide (3). Early and accurate prediction of DDIs is crucial for enhancing drug safety and improving patient outcomes. Proactively identifying potential interactions before clinical manifestation can prevent adverse drug events (ADEs), reduce hospital admissions, and lower the burden on healthcare systems (4). Predictive tools assist clinicians in making informed decisions during drug prescribing and facilitate regulatory agencies in evaluating drug safety profiles. Consequently, early DDI prediction plays an essential role in personalized medicine by optimizing therapeutic regimens tailored to individual patients’ medication profiles (5). Traditional approaches for DDI detection primarily rely on in vitro experiments, animal studies, and clinical trials. Although these methods provide valuable mechanistic insights, they are often time-consuming, costly, and limited in scope due to ethical and practical constraints (6). Post-marketing surveillance and spontaneous reporting systems capture real-world interaction data but suffer from underreporting and delayed identification of rare or complex interactions (7). Additionally, manual curation and rule-based systems, while effective for known DDIs, struggle to scale with the rapidly growing number of drugs and novel compounds, limiting their predictive capacity (8). The advent of artificial intelligence (AI) and machine learning (ML) has revolutionized healthcare by enabling data-driven, scalable, and efficient analysis of complex biomedical datasets (9). AI-based predictive modeling leverages large volumes of heterogeneous data—including chemical structures, biological pathways, electronic health records, and pharmacovigilance reports—to uncover hidden patterns and predict potential DDIs with greater accuracy (10). Machine learning algorithms, such as deep learning, support vector machines, and ensemble methods, have demonstrated promising results in modeling nonlinear interactions and integrating multi-modal data sources (11). These technologies not only enhance prediction accuracy but also offer interpretability and adaptability, making them powerful tools in drug safety assessment. This review aims to provide a comprehensive overview of AI-based predictive modeling approaches for drug-drug interactions. It explores the current state-of-the-art AI and ML techniques, the types of data utilized, and the challenges encountered in model development and validation. The review further discusses the integration of AI tools into clinical workflows and regulatory frameworks, emphasizing their potential to transform drug safety monitoring and personalized medicine. Finally, it highlights future directions and emerging trends in this rapidly evolving field.

Figure 1. Schematic representation of pharmacokinetic and pharmacodynamic processes involved in drug–drug interactions

Pharmacokinetic interactions influence the absorption, distribution, metabolism, and elimination (ADME) of drugs through mechanisms such as solubility changes, transporter modulation (e.g., OCT1, OCT3, OATP, P-gp, BCRP), and enzymatic metabolism (CYPs, UGTs). Pharmacodynamic interactions occur at the site of action or secondary tissues, affecting therapeutic response, adverse drug events, and overall clinical outcomes. The figure highlights the interplay between medicinal molecules, food products, and human physiology, emphasizing how these factors determine the therapeutic window, toxic effects, and treatment efficacy.

2. Drug-Drug Interactions: Concepts and Clinical Relevance

2.1 Types of DDIs: Pharmacokinetic, Pharmacodynamic, and Pharmaceutical

Pharmacokinetic interactions arise when one drug influences the absorption, distribution, metabolism, or excretion (ADME) of another drug, consequently altering its plasma concentration, bioavailability, or duration of action. These changes can lead to subtherapeutic levels or toxicity, depending on the direction of the interaction.

Absorption: Drug A may inhibit or enhance the gastrointestinal absorption of Drug B by altering pH levels, affecting transporter activity (e.g., P-glycoprotein), or forming insoluble complexes. For example, antacids can reduce the absorption of tetracyclines by forming chelates (12).

Distribution: Some drugs can displace others from plasma protein binding sites (e.g., albumin), leading to an increase in the free, active form of the displaced drug. An example is the displacement of warfarin by nonsteroidal anti-inflammatory drugs (NSAIDs), which may enhance anticoagulant activity and bleeding risk (13).

Metabolism: The cytochrome P450 (CYP) enzyme system is central to metabolic interactions. Inhibitors (e.g., ketoconazole) can increase the plasma levels of drugs metabolized by the same enzyme (e.g., midazolam), while inducers (e.g., rifampicin) can reduce their levels by enhancing clearance (14).

Excretion: Drugs may compete for renal tubular secretion or alter renal blood flow, affecting the elimination of co-administered agents. For instance, probenecid inhibits the renal excretion of penicillin, increasing its plasma concentration and duration of action. Pharmacokinetic DDIs are particularly significant because they can affect the dose-response relationship of drugs, requiring dosage adjustments to avoid toxicity or therapeutic failure (15).

Cytochrome P450 Enzyme System and Its Role in Drug Interactions

The cytochrome P450 (CYP450) enzyme system, predominantly located in the liver, plays a crucial role in the metabolism of a wide variety of drugs. It functions primarily by oxidizing xenobiotics, thereby enhancing their solubility and promoting excretion. However, this metabolic system is also a common site for drug-drug interactions, particularly through enzyme inhibition or enzyme induction, which can significantly alter the pharmacokinetics of co-administered drugs.

Figure 2. Metabolic processing of xenobiotics in hepatocytes via CYP-mediated oxidation, conjugation, and elimination, with pathways leading to detoxification or reactive metabolite formation.

The Figure 2, depicts the sequential metabolism of xenobiotics in hepatocytes, beginning with uptake via transporters, oxidation by CYP enzymes, conjugation by transferases, and eventual elimination through efflux transporters. It also shows how certain pathways generate reactive metabolites, contributing to drug–drug interactions and potential toxicities.

Enzyme Inhibition

Enzyme inhibition occurs when one drug inhibits the metabolic activity of a CYP450 isoenzyme responsible for the metabolism of another drug. This inhibition can lead to increased plasma concentrations of the affected drug, thereby enhancing its pharmacologic or toxic effects. For example, co-administration of ketoconazole, a potent inhibitor of CYP3A4, with drugs such as midazolam or simvastatin can result in elevated levels of the latter drugs, increasing the risk of adverse effects like respiratory depression or myopathy, respectively (16).

Enzyme Induction

Conversely, enzyme induction involves the upregulation of CYP450 enzymes, usually through increased gene transcription, resulting in enhanced metabolism of substrate drugs. This may lead to subtherapeutic drug levels and reduced clinical efficacy. For instance, rifampin, a strong inducer of CYP3A4, can lower plasma levels of drugs like oral contraceptives or antiretroviral agents, compromising their effectiveness (17).

Clinical Implications

Understanding these interactions is critical in clinical practice, as they can result in treatment failure, toxicity, or unpredictable therapeutic responses. Therefore, when prescribing multiple drugs metabolized by the CYP450 system, healthcare professionals must consider the potential for such interactions and adjust dosages or choose alternative therapies accordingly.

Pharmacodynamic Interactions

Pharmacodynamic interactions refer to the modifications in the effects of a drug due to the presence of another drug, without any change in their plasma concentrations or pharmacokinetic parameters such as absorption, distribution, metabolism, or excretion. These interactions arise from the drugs acting on the same or interconnected physiological systems, particularly at receptor sites, ion channels, enzymes, or cellular pathways (18).

Table 1. Classification and Examples of Pharmacodynamic Interactions

Type of Interaction

Definition

Example

Reference

Additive Interaction

Occurs when two drugs produce a combined effect equal to the sum of their individual effects, usually when they have similar pharmacological actions.

Co-administration of aspirin and acetaminophen producing additive analgesic effects via different pain pathways.

(19)

Synergistic Interaction

Occurs when the combined effect of two drugs is greater than the sum of their individual effects, often increasing therapeutic or adverse effects.

Concurrent use of benzodiazepines and opioids leading to profound CNS depression and life-threatening respiratory compromise.

(20)

Antagonistic Interaction

Occurs when the effect of one drug is reduced or inhibited by another drug, often through receptor competition or functional opposition.

Flumazenil reversing the sedative effects of benzodiazepines by acting as a competitive antagonist at GABA receptors.

(21)

Clinical Implications: Understanding pharmacodynamic interactions is essential in clinical practice, particularly in populations receiving multiple drugs (polypharmacy), such as the elderly or patients with chronic illnesses. Failure to recognize these interactions can lead to therapeutic failure, adverse drug reactions, or even fatal outcomes, especially in scenarios involving synergistic CNS depression.

Pharmaceutical Interactions: Pharmaceutical interactions, also known as pharmaceutical incompatibilities, occur prior to drug absorption, typically during the preparation, compounding, or administration phases of drug therapy. These interactions are non-biological in nature and arise from physical or chemical incompatibilities between drugs or excipients when mixed outside the body, especially in intravenous (IV) solutions or other dosage forms (22).

Mechanism of Pharmaceutical Interactions

Pharmaceutical interactions usually result from one or more of the following factors:

Physical incompatibility: This involves visible changes such as precipitation, turbidity, color changes, or gas formation. For example, mixing calcium-containing IV solutions with phosphate-containing solutions can result in the formation of insoluble calcium phosphate precipitates, which can be potentially harmful if administered intravenously (23).

Chemical incompatibility: Chemical reactions between drugs may lead to degradation, loss of potency, or formation of toxic by-products. Oxidation, hydrolysis, or acid-base reactions can alter drug stability. A notable example is the incompatibility between furosemide and acidic drugs like dopamine when mixed in the same IV line, which can lead to degradation of furosemide due to pH changes (24).

Clinical Implications: Although often preventable through proper preparation techniques and awareness of compatibility data, pharmaceutical interactions can have serious clinical consequences. These include reduced therapeutic efficacy, increased toxicity, and patient harm. Healthcare professionals must consult compatibility charts and drug references before mixing drugs, particularly in parenteral nutrition or intravenous therapy (25).

2.2 Real-World Examples and Statistics on Adverse Drug Events

Drug-drug interactions (DDIs) represent a significant clinical concern as they are a major cause of adverse drug events (ADEs), which contribute substantially to patient morbidity and mortality worldwide. ADEs resulting from DDIs can range from mild discomfort to life-threatening conditions, making them a critical focus in patient safety and pharmacovigilance.

Prevalence of ADEs Related to DDIs

It is estimated that 6 to 30% of all hospital admissions are related to adverse drug events, with a considerable proportion of these events directly attributable to drug-drug interactions. This wide range reflects differences in patient populations, healthcare settings, and methods of detection but highlights the pervasive impact of DDIs in clinical practice (26).

Clinical Consequences

The consequences of DDIs-induced ADEs include prolonged hospital stays, increased healthcare costs, and elevated risk of morbidity and mortality. Vulnerable populations such as the elderly, patients on multiple medications (polypharmacy), and those with chronic illnesses are particularly at risk. Effective identification and management of potential drug-drug interactions (DDIs) are critical steps in minimizing preventable adverse drug events (ADEs) and enhancing therapeutic outcomes. Failure to recognize clinically significant DDIs can lead to serious complications, increased hospital admissions, and even mortality (27).

Clinical Example: Warfarin and NSAIDs Interaction

A well-documented example involves the concomitant use of warfarin, an oral anticoagulant, with nonsteroidal anti-inflammatory drugs (NSAIDs). This combination significantly elevates the risk of bleeding due to the additive effects of NSAIDs on platelet function and gastrointestinal mucosa, alongside warfarin’s anticoagulant properties. Such interactions have been linked to numerous emergency hospital visits, emphasizing the need for careful drug selection, monitoring, and patient education to prevent adverse outcomes. Another notable example is the co-administration of statins with certain antibiotics like clarithromycin, which can elevate statin levels and precipitate severe muscle toxicity (rhabdomyolysis). The presence of drug-drug interactions (DDIs) underscores the critical necessity for accurate prediction and effective prevention strategies to mitigate associated risks, particularly in patients undergoing polypharmacy. Polypharmacy, defined as the concurrent use of multiple medications, is especially prevalent among elderly populations who often have multiple comorbidities requiring complex therapeutic regimens (28).

Vulnerability of Elderly Populations

Elderly patients are more susceptible to adverse outcomes from DDIs due to age-related physiological changes such as altered pharmacokinetics and pharmacodynamics, as well as the increased likelihood of receiving multiple medications. The complexity of their medication regimens heightens the risk of harmful interactions, making it imperative to implement robust screening tools, medication reviews, and clinical decision support systems to predict and prevent DDIs. Such proactive approaches can significantly reduce preventable adverse drug events and improve patient safety in this vulnerable group (29).

2.3 Regulatory Perspectives on DDIs

Regulatory agencies, including the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA), play a pivotal role in managing drug-drug interactions (DDIs) throughout the drug development lifecycle and post-marketing surveillance.

FDA Recommendations for DDI Assessment

The FDA has established comprehensive guidelines that mandate systematic evaluation of new drugs for their potential to cause clinically significant DDIs. This process typically begins with in vitro testing to determine whether a drug can inhibit or induce key drug-metabolizing enzymes, especially cytochrome P450 (CYP) isoforms, or affect drug transporters such as P-glycoprotein. Based on these initial findings, the FDA recommends conducting in vivo pharmacokinetic studies to assess the magnitude of these interactions in humans. Clinical DDI studies are performed as necessary to guide safe drug labeling and dosing recommendations, thereby ensuring patient safety and optimizing therapeutic efficacy. Additionally, drug labels must clearly describe known interactions and provide dosing recommendations to healthcare providers. Regulatory frameworks are increasingly acknowledging the value of computational methods, including artificial intelligence (AI)-based predictive models, as complementary tools in the assessment of drug-drug interaction (DDI) risks. These innovative approaches utilize large datasets, machine learning algorithms, and molecular modeling to predict potential DDIs early in the drug development process (30).

Benefits of AI-Based Predictive Models

AI-driven models enhance the capacity to identify interaction risks before clinical exposure, thus accelerating decision-making and optimizing the design of in vitro and in vivo studies. Furthermore, they support post-marketing drug safety monitoring by enabling continuous evaluation of real-world data and identifying novel or rare interactions that may not be evident during clinical trials. The integration of these computational tools into regulatory assessments helps improve the overall safety profile of drugs and informs evidence-based regulatory decisions (31).

3. Conventional Approaches for DDI Detection And Prediction

3.1 Experimental Approaches: In Vitro and In Vivo Studies

Conventional Methods for Detecting and Predicting Drug-Drug Interactions

The detection and prediction of drug-drug interactions (DDIs) predominantly depend on experimental methodologies, which include both in vitro and in vivo studies designed to evaluate the interaction potential of candidate drugs.

In Vitro Studies

In vi/tro approaches are widely employed in the early phases of drug development to assess whether a drug can inhibit or induce key drug-metabolizing enzymes and transport proteins. These studies commonly utilize biological preparations such as:

3.1.1 Human Liver Microsomes: These subcellular fractions contain cytochrome P450 enzymes and other metabolic components, allowing the evaluation of a drug’s effect on enzyme activity under controlled conditions.

3.1.2 Recombinant Enzymes: Purified individual enzymes expressed in vitro are used to identify specific CYP isoforms involved in drug metabolism and to characterize inhibition or induction profiles.

3.1.3 Cell Cultures: Cultured human hepatocytes or other relevant cell lines provide a more physiologically relevant system to study enzyme induction and transporter activity, capturing complex regulatory mechanisms that influence drug metabolism (32).These assays primarily focus on the cytochrome P450 (CYP) family, which is responsible for the metabolism of the majority of drugs, making it critical to assess the potential modulation of these enzymes to predict DDIs that could impact drug safety and efficacy.In vivo studies play a crucial role in the comprehensive evaluation of drug-drug interactions (DDIs) by utilizing animal models and human clinical trials to investigate pharmacokinetic and pharmacodynamic interactions within the context of whole-organism physiological conditions . These studies serve to validate and complement findings from in vitro experiments, as they account for the complexities of drug absorption, distribution, metabolism, and excretion (ADME), as well as receptor-mediated effects that cannot be fully replicated in isolated systems (33).

Animal Models

Animal studies provide preliminary insight into the potential DDIs by examining how drugs interact in vivo, considering factors such as enzyme induction or inhibition, transporter effects, and systemic physiological responses. These models help identify interactions that might affect drug bioavailability and toxicity before advancing to human trials.

Human Clinical Trials

Clinical drug interaction studies, often designed as crossover or parallel group trials, are fundamental to confirm the presence and clinical significance of DDIs observed in preclinical models. These trials assess alterations in pharmacokinetic parameters, such as plasma drug concentrations and clearance, and pharmacodynamic outcomes, including efficacy and adverse effects. The results inform necessary dosing adjustments, precautions, or contraindications to ensure patient safety and optimal therapeutic effect (34).

3.2 Rule-Based and Knowledge-Based Systems

In addition to traditional experimental methods, computational approaches have been increasingly developed and integrated to enhance the prediction of drug-drug interactions (DDIs). Among these, rule-based and knowledge-based systems are prominent tools that leverage pharmacological principles and expert knowledge to identify potential interaction risks.

Rule-Based Systems

Rule-based systems operate by applying logical “if-then” decision rules derived from established biochemical and pharmacological data. These rules encode relationships such as enzyme inhibition or induction, substrate specificity, and receptor binding. For example, a typical rule might state: if Drug A is a known inhibitor of cytochrome P450 enzyme CYP3A4 and Drug B is predominantly metabolized by CYP3A4, then a significant interaction is likely to occur (35). By systematically applying such rules, the system can flag probable interactions before clinical exposure.

Knowledge-Based Databases

These computational systems rely heavily on curated databases that compile comprehensive information, including metabolic enzyme pathways, transporter substrates, receptor targets, and documented adverse events reported in the literature or pharmacovigilance databases. The integration of this extensive data enables accurate and evidence-based prediction of DDIs, facilitating early risk assessment and informed decision-making in drug development and clinical practice (36).

Knowledge-Based Systems in DDI Prediction

Knowledge-based systems build upon traditional rule-based frameworks by incorporating a wider array of pharmacological data to enhance the accuracy and scope of drug-drug interaction (DDI) predictions. These systems integrate information on drug chemical properties, therapeutic classes, and documented adverse event profiles, enabling a more comprehensive evaluation of both pharmacokinetic and pharmacodynamic interactions (37).

Comprehensive Data Integration

By combining diverse datasets, knowledge-based systems can identify complex interaction mechanisms that may not be evident through simple rule-based logic alone. This holistic approach allows for the prediction of interactions involving multiple pathways, including enzyme metabolism, drug transporters, receptor binding, and downstream physiological effects (38).

Clinical and Regulatory Utility

Moreover, the output generated by these systems is typically interpretable and transparent, which facilitates their integration into clinical decision support tools. This supports healthcare professionals in making informed prescribing decisions and assists regulatory agencies during drug evaluation and post-marketing surveillance to ensure drug safety (39).

3.3 Limitations: Cost, Time, Scalability, and Generalizability

Despite the widespread use and clinical relevance of conventional experimental and computational approaches in predicting drug-drug interactions (DDIs), each method has inherent limitations that restrict its scalability, efficiency, and predictive accuracy.

Limitations of Experimental Methods

Traditional experimental methods, including in vitro assays, animal models, and clinical trials, are considered the gold standard for DDI detection. However, these methods are often resource-intensive, requiring substantial financial investment, specialized personnel, and extended timelines to complete laboratory work and human studies. Moreover, ethical concerns limit the extent of in vivo testing, particularly when evaluating potentially hazardous drug combinations, making it challenging to explore certain interaction scenarios during development (40). A critical challenge lies in the scalability of these approaches. With the growing number of approved pharmaceutical agents, the number of possible pairwise drug combinations increases exponentially. Testing each possible combination using traditional methods is practically unfeasible, resulting in many clinically relevant DDIs being unidentified during pre-marketing phases (41).

Limitations of Rule-Based and Knowledge-Based Systems

Although rule-based and knowledge-based systems offer a more efficient and scalable alternative, their performance is largely dependent on the quality, completeness, and timeliness of existing pharmacological data. These systems rely on predefined rules and curated databases, which may not include emerging, rare, or novel DDIs, thus limiting their ability to detect previously unreported interactions. Furthermore, these systems often lack generalizability across diverse patient populations and drug classes because they do not adapt dynamically to new data inputs. Unlike data-driven models, they do not learn or improve over time, making them less suitable for real-time surveillance or for use in personalized medicine settings (42).

Need for Advanced Computational Solutions

These limitations underscore the urgent need for advanced, data-driven approaches, such as artificial intelligence (AI) and machine learning (ML) models, which can integrate and analyze large-scale, heterogeneous datasets. Such technologies offer the potential to overcome current predictive gaps, improve accuracy, and enhance the detection of both known and previously unidentified DDIs.

4. AI In Drug Discovery And Ddi Prediction

4.1 Overview of AI/ML Techniques in Biomedical Applications

Artificial intelligence (AI) and machine learning (ML) have emerged as transformative technologies in biomedical research, offering powerful tools to enhance drug discovery, development, and safety assessment processes. AI encompasses a broad range of computational methods that allow machines to learn from large datasets, identify patterns, and make informed decisions with minimal human intervention (43).

Machine Learning Algorithms in Biomedical Applications

Table 2. Commonly Used Machine Learning Algorithms in Biomedical Applications

Machine Learning Algorithm

Key Applications in Biomedical Domain

Support Vector Machines (SVM)

Classification tasks in drug–target interaction prediction and toxicity modeling.

Random Forests

Robust feature selection and ensemble learning; used in pharmacovigilance and adverse event prediction.

Deep Neural Networks (DNNs)

Modeling non-linear relationships in high-dimensional data; effective for large-scale compound screening and multi-drug interaction prediction.

Natural Language Processing (NLP) Models

Extraction of relevant information from scientific literature, clinical notes, and adverse event reports to identify potential drug–drug interactions (DDIs) and drug safety signals (44).

Applications in Drug Development and Safety

These AI-driven techniques facilitate a wide array of applications across the drug development pipeline, including target identification, virtual compound screening, and prediction of pharmacokinetic and pharmacodynamic properties. Notably, AI and ML have shown considerable promise in predicting drug efficacy, toxicity, and drug-drug interactions (DDIs) by integrating heterogeneous data sources such as chemical structures, omics data, and electronic health records (45).

4.2 Advantages of AI in Handling Complex, High-Dimensional Data

One of the fundamental strengths of artificial intelligence (AI) lies in its capacity to process and analyze large-scale, complex, and high-dimensional datasets, which are characteristic of modern biomedical research. These datasets commonly include a broad spectrum of information such as chemical structures, genomic profiles, transcriptomic and proteomic data, electronic health records (EHRs), and pharmacovigilance reports. The heterogeneous and voluminous nature of such data often presents significant challenges for traditional statistical techniques, which typically require structured input formats and may struggle to model non-linear or multi-dimensional relationships effectively (46).

AI Capabilities in Modeling Complex Interactions

AI algorithms—particularly those employing deep learning architectures—are capable of identifying nonlinear patterns, hidden correlations, and latent features within the data without the need for extensive manual feature engineering. This flexibility makes them especially suitable for drug-drug interaction (DDI) prediction, where complex interdependencies between molecular, clinical, and patient-level variables must be considered simultaneously. In this context, AI models can integrate diverse data modalities, such as molecular fingerprints, metabolic pathways, drug targets, and patient-specific characteristics (e.g., age, gender, genetic polymorphisms), allowing for comprehensive modeling of both pharmacokinetic and pharmacodynamic interactions (47).

Continuous Learning and Adaptability

Moreover, a distinct advantage of AI lies in its ability to learn and adapt continuously. As new data become available—through clinical trials, pharmacovigilance systems, or real-world evidence—AI models can be retrained or fine-tuned to reflect emerging knowledge and novel drug combinations, thereby enhancing their predictive accuracy, generalizability, and relevance in clinical and regulatory settings.

4.3 General Workflow of AI-Based Predictive Modeling

The development of AI-based predictive models for drug-drug interaction (DDI) detection follows a structured, multi-phase workflow that spans from data acquisition to model deployment and interpretation.

Data Collection and Preprocessing

The first stage involves the collection and preprocessing of relevant datasets from a wide variety of biomedical sources. These include chemical structure databases (e.g., PubChem, ChEMBL), biological assay results, clinical data repositories, and scientific literature. The raw data are then subjected to cleaning, normalization, and integration, ensuring consistency and compatibility across heterogeneous sources. During this stage, feature extraction is performed to transform raw data into a format suitable for machine learning. Features may consist of chemical descriptors, drug-target interaction profiles, enzyme activity data, or patient demographics (48).

Model Training and Learning Strategies

Once the data are structured and features are engineered, the next phase involves training machine learning models. In supervised learning approaches, the model is trained on labeled datasets where DDIs are already known, allowing it to learn patterns that differentiate interacting from non-interacting drug pairs. Alternatively, unsupervised learning techniques are employed when labels are unavailable, aiming to discover novel clusters or latent relationships that may indicate potential interactions. Popular algorithms used in this context include support vector machines (SVMs), random forests, graph neural networks, and deep learning models such as convolutional neural networks (CNNs) and recurrent neural networks (RNNs). To ensure model robustness and generalizability, cross-validation techniques—such as k-fold validation—are implemented during training to assess performance and prevent overfitting (49).

Prediction, Interpretation, and Application

After successful training, the validated model is deployed to predict DDIs in previously uncharacterized drug pairs. These predictions are typically expressed as probabilistic interaction scores or binary/multiclass classification outputs. Given the complexity of AI models, especially deep learning architectures, interpretability techniques—such as SHAP (Shapley Additive Explanations), LIME (Local Interpretable Model-agnostic Explanations), or attention mechanisms—are often applied to explain model outputs. This interpretability supports clinical decision-making, enhances regulatory transparency, and increases trust in AI-generated predictions.

5. Data Sources For AI-Based Ddi Modeling

5.1 Structured Databases

The effectiveness of AI-based predictive modeling for drug-drug interactions (DDIs) is fundamentally dependent on the availability and quality of structured, high-confidence biomedical databases. These curated resources serve as the foundational backbone for data-driven models by providing comprehensive information on drug properties, molecular targets, interaction mechanisms, and adverse event profiles. They are instrumental in both training machine learning models and validating predictive outputs.

5.1.1 DrugBank

One of the most extensively used databases in this domain is DrugBank, which offers richly annotated entries for thousands of approved and investigational drugs. The database includes detailed chemical structures, pharmacokinetics, mechanisms of action, drug–target interactions, and pharmaceutical data, making it a valuable resource for constructing input features for AI models (50).

5.1.2 KEGG (Kyoto Encyclopedia of Genes and Genomes)

The KEGG database links drugs to their associated biological pathways, genes, and cellular processes, facilitating mechanistic understanding of DDIs. By providing insights into metabolic pathways, signal transduction, and disease associations, KEGG enables the mapping of drug interactions to specific biological systems, thereby enriching the interpretability of AI predictions (51).

5.1.3 TWOSIDES

The TWOSIDES database compiles statistically inferred adverse effects resulting from drug combinations, derived from large-scale post-marketing surveillance and electronic health record (EHR) data. This allows for the identification of unexpected or emergent DDIs that may not be evident from clinical trials alone, offering real-world context for model development.

5.1.4 SIDER (Side Effect Resource)

The SIDER database aggregates information on known adverse drug reactions (ADRs) associated with marketed pharmaceuticals. This resource is critical for linking DDIs to clinical outcomes, helping AI systems learn not just the molecular basis of interactions but also their real-world consequences.

5.1.5 ChEMBL

ChEMBL provides bioactivity data for a wide range of drug-like molecules and their biological targets, compiled from medicinal chemistry literature. The database supports the prediction of DDIs at the molecular interaction level, particularly through quantitative structure-activity relationships (QSAR) and ligand–target modelling. Together, these structured databases form the informational infrastructure required for robust AI model development, offering curated and diverse data essential for predicting, validating, and interpreting DDIs (52).

5.2 Real-World Data: Electronic Health Records (EHRs) and Adverse Event Reports

In addition to structured and curated biomedical databases, the integration of real-world data (RWD) sources—such as Electronic Health Records (EHRs) and spontaneous adverse event reporting systems—provides critical, patient-specific insights that enhance the depth and applicability of AI-based drug-drug interaction (DDI) prediction models.

5.2.1 Electronic Health Records (EHRs)

EHRs offer comprehensive, longitudinal clinical data, including medication prescriptions, laboratory results, diagnostic information, comorbidities, and treatment outcomes. This real-time, patient-centered data enables AI algorithms to examine drug combinations as they are administered across diverse populations under real-world clinical conditions. Unlike controlled clinical trials, EHRs reflect the heterogeneity of routine medical practice, making them especially valuable for uncovering context-dependent or rare DDIs. For example, certain interactions may only become apparent in subgroups defined by age, renal function, or polypharmacy status—scenarios that are often underrepresented in clinical trials (53).

5.2.2 Spontaneous Adverse Event Reporting Systems

Spontaneous reporting systems, such as the U.S. Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS), provide an extensive repository of post-marketing drug safety data. These systems compile voluntarily reported cases of adverse drug events (ADEs) from healthcare professionals, patients, and manufacturers. Mining FAERS using AI and statistical signal detection methods can uncover previously unrecognized DDIs, especially those associated with unexpected or rare adverse outcomes. Such insights are instrumental in identifying emerging drug safety concerns and informing regulatory actions (54).

5.2.3 Limitations of Real-World Data Sources

Despite their importance, real-world data sources pose several methodological challenges. For instance, underreporting is common in spontaneous reporting systems, particularly for non-serious or delayed adverse events. Additionally, reporting biases—such as stimulated reporting following media attention or litigation can distort the perceived frequency or severity of specific DDIs. In the context of EHRs, data heterogeneity across healthcare systems and inconsistent documentation practices can compromise the completeness, standardization, and interpretability of the data. Therefore, rigorous data preprocessing and the application of robust AI methods are essential to extract reliable and clinically meaningful insights from these sources. Nevertheless, when effectively leveraged, real-world data serves as a critical supplement to traditional datasets, enhancing the clinical relevance, sensitivity, and generalizability of AI-driven DDI prediction frameworks (55).

5.3 Literature Mining

In addition to structured databases and real-world clinical data, biomedical literature serves as a critical and dynamic source of information for enhancing the scope and accuracy of AI-based drug-drug interaction (DDI) prediction models. Vast repositories such as PubMed, MEDLINE, and other scholarly databases contain a wealth of unstructured textual data encompassing case reports, clinical trial outcomes, mechanistic pharmacology studies, and drug safety communications that often detail newly identified or rare DDIs (56).

5.3.1 Role of Automated Literature Mining

To efficiently process this expansive body of literature, automated text mining techniques are employed to extract DDI-relevant information. These systems scan and parse millions of articles to identify explicitly reported drug interactions, as well as contextual clues about drug mechanisms, enzyme modulation, and clinical outcomes. This allows researchers to incorporate the most current evidence into their predictive frameworks—particularly valuable in cases where novel drug combinations or emerging safety concerns have not yet been encoded in structured databases (57).

5.3.2 Application of Natural Language Processing (NLP)

Natural Language Processing (NLP) algorithms are integral to this process. These algorithms are designed to interpret unstructured biomedical text, enabling the identification and extraction of key entities such as drug names, target enzymes (e.g., CYP450 isoforms), transporters, and adverse drug reactions. More importantly, NLP models establish semantic relationships between these entities—such as inhibition, induction, or causality—thereby contributing structured, machine-readable data from free-text sources. Advanced NLP frameworks, including named entity recognition (NER), relation extraction, and dependency parsing, enable the construction of knowledge graphs or interaction networks that can be used directly in machine learning models for DDI prediction. These tools not only enrich existing datasets but also ensure that AI systems are continuously updated with the latest scientific discoveries and regulatory alerts.Thus, biomedical literature mining—powered by NLP—represents a vital strategy for bridging the gap between published scientific knowledge and computational DDI modeling, significantly enhancing both predictive coverage and clinical relevance(58).

5.4 Challenges in Data Quality, Integration, and Standardization

Despite the increasing availability of diverse and rich data sources for drug-drug interaction (DDI) modeling, numerous technical, methodological, and regulatory challenges hinder their effective integration into AI-based predictive frameworks. These limitations span across data quality, interoperability, and privacy concerns, all of which must be addressed to fully realize the potential of AI in pharmacovigilance and drug safety.

5.4.1 Variability and Limitations in Data Quality

The quality and completeness of available data sources vary significantly. While structured biomedical databases (e.g., Drug Bank, ChEMBL) are extensively curated, they may still contain outdated, incomplete, or infrequently updated information that can limit the predictive capacity of AI models. In contrast, real-world data (RWD)—including electronic health records (EHRs) and adverse event reporting systems—is often noisy, inconsistent, and subject to various biases, such as underreporting or selective documentation of adverse outcomes. These quality issues can propagate through AI models, potentially reducing their accuracy and increasing the risk of false predictions (59).

5.4.2 Data Integration and Harmonization Challenges

AI-based DDI prediction requires the integration of heterogeneous data types, including molecular structures, genomic and proteomic profiles, clinical records, and textual literature. However, these data sources are typically formatted using incompatible terminologies, structures, and ontologies, making data harmonization a major challenge. Mapping disparate data into a unified framework necessitates the application of standardized vocabularies and ontologies, such as RxNorm, SNOMED CT, and the Unified Medical Language System (UMLS). Without these standardization efforts, AI systems may struggle to interpret cross-domain inputs consistently or exchange information between platforms.

5.4.3 Interoperability and Privacy Constraints

The lack of interoperability among healthcare databases and AI platforms further complicates collaborative research and model deployment. Moreover, access to patient-level dataparticularly within EHRs—is often restricted due to privacy regulations, including HIPAA (Health Insurance Portability and Accountability Act) in the U.S. and GDPR (General Data Protection Regulation) in the EU. These frameworks impose strict data governance requirements, limiting the free exchange of sensitive health information for machine learning applications. To overcome these barriers, advanced methodologies such as federated learning have been introduced. Federated learning enables decentralized model training, allowing multiple institutions to collaboratively build AI models without sharing raw patient data, thereby maintaining data privacy and compliance (60).

5.4.4 The Need for Systemic Solutions

Addressing these challenges requires a multi-faceted strategy involving ongoing efforts in data curation, standardization, privacy-preserving technologies, and regulatory reform. Without tackling these foundational issues, the potential of AI to revolutionize DDI prediction and advance personalized medicine and drug safety monitoring will remain unrealized.

6. AI and Machine Learning Techniques For Ddi Prediction

6.1 Traditional Machine Learning

Traditional machine learning (ML) algorithms have been extensively employed in the prediction of drug-drug interactions (DDIs) due to their proven ability to model complex relationships within structured datasets. These methods typically require explicit feature engineering and domain knowledge to extract meaningful attributes from raw data, such as chemical structures, biological properties, and phenotypic effects.

Figure 3. Workflow of Random Forest–based predictive modeling for drug–drug interaction (DDI) prediction.

This schematic illustrates a Random Forest (RF) workflow integrating both experimental and computational approaches for DDI prediction. Initially, peptide libraries and receptor structures undergo experimental affinity testing and docking-based virtual screening to generate actual and predicted interaction data. These datasets are merged and processed to train the RF model, enabling it to learn patterns that distinguish interacting from non-interacting pairs. Once trained, the model is applied to new, untested drug data, producing predictions with interaction classes, confidence scores, and potential binding profiles. This approach demonstrates the effectiveness of combining heterogeneous datasets in traditional machine learning for accurate DDI prediction.

Logistic regression is a widely used statistical model for binary classification tasks and serves as a baseline method for DDI prediction. It estimates the probability of interaction between drug pairs by fitting a linear combination of features through a logistic function. Despite its simplicity, logistic regression provides interpretable coefficients that help understand feature importance, but it may struggle with capturing nonlinear dependencies inherent in biological systems.

Decision trees offer a rule-based approach by recursively partitioning the feature space into subsets based on threshold values, forming a tree-like model that classifies drug pairs as interacting or non-interacting. Their intuitive structure allows for easy interpretation, but single trees tend to overfit, limiting their generalization. To overcome overfitting and improve predictive accuracy, ensemble methods such as random forests aggregate multiple decision trees trained on bootstrapped samples of data and random subsets of features. This approach reduces variance and captures complex, nonlinear relationships between features, making random forests a popular choice for DDI prediction tasks (61) .

Support Vector Machines (SVMs) are powerful classifiers that identify the optimal hyperplane separating interacting from non-interacting drug pairs in a high-dimensional feature space. With kernel functions, SVMs can model nonlinear interactions and handle heterogeneous data types, including chemical fingerprints and biological annotations. However, SVMs require careful parameter tuning and may be computationally intensive with large datasets.Feature engineering is critical to the success of traditional ML models in DDI prediction. Features are typically derived from multiple domains:

Chemical data such as molecular fingerprints, physicochemical properties, and structural descriptors represent the intrinsic properties of drugs that influence interaction potential.

Biological data include information on drug targets, enzymes, transporters, and pathways, which help model pharmacokinetic and pharmacodynamic mechanisms underlying DDIs.

Phenotypic data capture observable effects such as side effects, therapeutic classes, and clinical outcomes, providing contextual clues for potential interactions. Careful integration and selection of these features allow traditional ML algorithms to effectively discriminate between interacting and non-interacting drug pairs, contributing to early identification and risk mitigation in clinical practice (62).

6.2 Deep Learning Approaches

Deep learning, a subset of machine learning, has gained significant traction in drug-drug interaction (DDI) prediction due to its ability to automatically learn complex, hierarchical representations from raw data without extensive manual feature engineering. Deep learning models can capture intricate nonlinear relationships present in biological and chemical data, making them particularly suitable for modeling DDIs.

Feedforward Neural Networks (FNNs) are among the simplest deep learning architectures, consisting of multiple layers of interconnected neurons where information flows in one direction—from input to output. FNNs can model complex functions mapping drug features to interaction likelihoods. Despite their effectiveness, FNNs require carefully designed input features and may struggle to handle sequential or spatial dependencies inherent in molecular data.To address spatial relationships in drug structures, Convolutional Neural Networks (CNNs) have been employed. CNNs utilize convolutional layers to detect local patterns and motifs within molecular representations such as graphs, images, or fingerprints. By learning filters that capture structural subcomponents of drugs, CNNs improve the accuracy of DDI prediction by effectively encoding chemical properties and interaction-relevant features. For example, CNNs have been applied to molecular graphs where nodes represent atoms and edges represent chemical bonds, allowing the network to understand the spatial topology of molecules (63).

Recurrent Neural Networks (RNNs), including variants like Long Short-Term Memory (LSTM) and Gated Recurrent Units (GRU), are designed to model sequential data, making them suitable for analyzing drug-related sequences such as SMILES strings, amino acid chains, or time-series clinical data. RNNs capture temporal dependencies and contextual information, which are crucial when drug interactions depend on dynamic processes like metabolism or temporal co-administration. Additionally, autoencoders—unsupervised deep learning models—have been used for dimensionality reduction and feature extraction in DDI prediction. Autoencoders learn compressed, latent representations (deep embeddings) of input data by encoding and then reconstructing it, thus distilling essential features from high-dimensional molecular or biological data. These embeddings serve as informative inputs to downstream predictive models, enhancing their performance and generalizability. Together, these deep learning architectures have advanced the field of DDI prediction by enabling end-to-end modeling pipelines that learn from raw, heterogeneous data sources, capturing complex biological and chemical interactions beyond the reach of traditional methods (64).

6.3 Graph-Based Models

Graph-based models have emerged as powerful tools for drug-drug interaction (DDI) prediction by naturally representing complex biological and chemical relationships as networks. These models leverage the inherent graph structure of molecular entities, biological pathways, and interaction networks to capture rich contextual information often lost in traditional vector-based methods.

Graph Neural Networks (GNNs) have gained prominence in DDI prediction due to their ability to learn meaningful node and edge representations directly from graph-structured data. In this context, nodes typically represent drugs, targets, or proteins, while edges encode various types of relationships such as chemical similarity, physical interactions, or known DDIs. GNNs iteratively aggregate and transform information from a node’s neighbors, enabling the model to capture both local and global network topology and thereby predict novel interactions with high accuracy. Several studies have demonstrated that GNNs outperform classical machine learning models by effectively modeling the non-Euclidean structure of pharmacological data. Beyond GNNs, knowledge graphs integrate heterogeneous biomedical data, including drugs, diseases, genes, and pathways, into a unified graph framework. This approach allows AI models to exploit multi-relational data to understand mechanistic links underlying DDIs. Knowledge graphs facilitate the application of link prediction algorithms, which infer missing edges—potential interactions—based on observed network patterns and node attributes. Techniques such as graph embedding and matrix factorization are frequently used to generate low-dimensional vector representations that preserve network semantics, supporting robust interaction prediction (65).

Network pharmacology principles align well with graph-based modeling by emphasizing the complex interplay between multiple drugs, targets, and biological systems. Network-based link prediction methods utilize topological features such as common neighbors, shortest paths, and clustering coefficients to estimate the likelihood of interactions. These methods enable the identification of polypharmacology effects and synergistic or antagonistic drug combinations, aiding in precision medicine and adverse event prevention. Despite their advantages, graph-based approaches require high-quality, comprehensive interaction networks and careful handling of data sparsity and noise. Integration of diverse data sources into consistent graph formats and interpretability of complex network models remain ongoing challenges. Overall, graph-based models, particularly GNNs and knowledge graphs, represent a promising frontier in AI-driven DDI prediction, offering deeper insights into the molecular and systemic bases of drug interactions (66).

Figure 4: Workflow of the Know DDI framework combining DDI and biomedical knowledge graphs to generate embeddings, extract subgraphs, and classify predicted DDI types.

This schematic illustrates a graph-based AI approach for predicting drug–drug interactions by merging DDI graphs with knowledge graphs containing biomedical entities. Through embedding generation, subgraph extraction, and feature integration, the model captures both structural and semantic relationships between drugs, enabling accurate prediction of interaction types.

6.4 Natural Language Processing (NLP)

Natural Language Processing (NLP) plays a crucial role in the prediction and discovery of drug-drug interactions (DDIs) by enabling the automated extraction of relevant information from vast amounts of unstructured textual data. Scientific literature, clinical notes, electronic health records (EHRs), and adverse event reports contain valuable insights about DDIs that are often buried within complex narratives, making NLP indispensable for efficient knowledge extraction. A primary NLP application in DDI prediction is text mining, which involves identifying, retrieving, and structuring information related to drug interactions from heterogeneous text sources. Text mining techniques can process millions of biomedical articles and clinical documents to uncover previously unreported or poorly characterized interactions, thereby enriching existing DDI databases and supporting clinical decision-making (67) Key tasks within NLP for DDI extraction include Named Entity Recognition (NER) and Relation Extraction (RE). NER focuses on identifying and classifying drug names, dosages, adverse events, and other biomedical entities within text. Accurate recognition of these entities is fundamental for subsequent analysis and linking to structured databases. Following entity recognition, Relation Extraction (RE) aims to detect and characterize the relationships between identified entities—specifically, interactions between drugs. RE models utilize rule-based, supervised, or deep learning approaches to determine whether two or more drugs mentioned in a text have a potential interaction, along with the nature and severity of the interaction. Recent advances have leveraged transformer-based architectures, such as BERT and its biomedical adaptations, to significantly improve the precision and recall of DDI extraction from complex sentences. NLP techniques face challenges including ambiguous terminology, diverse writing styles, and incomplete reporting in clinical texts. Nonetheless, combining NLP with AI-driven predictive modeling enhances the comprehensiveness and accuracy of DDI prediction frameworks by integrating knowledge from both structured databases and unstructured textual data (68).

7. Model Evaluation and Validation Strategies

The development of robust and reliable AI-based models for drug-drug interaction (DDI) prediction demands rigorous evaluation and validation to ensure performance, generalizability, and clinical relevance. Evaluation not only helps identify model strengths and limitations but also guides model selection and optimization for real-world deployment. The assessment of DDI models typically involves quantitative performance metrics, validation techniques, external benchmarking, and growing concerns regarding interpretability and explainability.

7.1 Common Evaluation Metrics

o effectively evaluate the performance of drug-drug interaction (DDI) predictive models, a variety of statistical metrics are utilized. These metrics provide comprehensive insights into different aspects of model accuracy, reliability, and discriminative power, which are critical for both model comparison and optimization during training and validation phases.

7.1.1 Accuracy

Accuracy represents the proportion of total correct predictions made by the model, encompassing both true positive and true negative outcomes. While accuracy provides a straightforward measure of overall correctness, it can be misleading in the context of DDI prediction due to the common issue of dataset imbalance. Specifically, since non-interacting drug pairs vastly outnumber interacting pairs, a model that predominantly predicts no interaction can achieve high accuracy without effectively identifying true interactions (69).

7.1.2 Precision

Precision quantifies the ratio of correctly predicted positive interactions to all predicted positive instances. This metric reflects the model’s capacity to minimize false positives, thereby indicating how reliably the model identifies actual interactions among the predicted ones. High precision is especially valuable in clinical settings to avoid unnecessary alerts or interventions based on incorrect interaction predictions.

7.1.3 Recall (Sensitivity)

Recall, also known as sensitivity, measures the proportion of actual interacting drug pairs that the model successfully identifies. It assesses the model’s ability to detect true DDIs, which is essential for ensuring potentially harmful interactions are not overlooked. A high recall reduces the risk of false negatives, which is crucial for patient safety and effective pharmacovigilance.

7.1.4 F1-Score

The F1-score is the harmonic mean of precision and recall, providing a balanced evaluation metric that accounts for both false positives and false negatives. This measure is particularly useful in datasets with class imbalance, such as those encountered in DDI prediction, where it offers a more nuanced assessment of model performance compared to accuracy alone.

7.1.5 Area Under the Receiver Operating Characteristic Curve (ROC-AUC)

The ROC-AUC metric evaluates the model’s ability to discriminate between interacting and non-interacting drug pairs across all classification thresholds. By plotting the true positive rate against the false positive rate, ROC-AUC summarizes the trade-offs between sensitivity and specificity. This metric is widely adopted in biomedical binary classification tasks due to its robustness and interpretability, with higher values indicating superior discriminative performance. Collectively, these metrics form an essential toolkit for assessing, comparing, and refining AI models designed for DDI prediction, thereby enhancing their clinical relevance and reliability (70).

7.2 Cross-Validation Techniques

Cross-validation represents a fundamental methodology for assessing the performance of predictive models, particularly in scenarios where labeled datasets are limited, as is often the case in drug-drug interaction (DDI) research. This technique helps to prevent overfitting by providing an unbiased evaluation of the model's generalizability to unseen data.

7.2.1 K-Fold Cross-Validation

The most widely used form of cross-validation is k-fold cross-validation. In this approach, the entire dataset is randomly partitioned into k approximately equal-sized subsets, known as folds. During each iteration, the model is trained on k–1 folds and subsequently tested on the remaining single fold. This process is repeated k times, ensuring that each fold serves as the test set exactly once. By averaging performance metrics over the k iterations, this method yields a more reliable and stable estimate of model effectiveness compared to a single train-test split (71).

7.2.2 Stratified Cross-Validation

When dealing with DDI datasets, which typically exhibit significant class imbalance between interacting and non-interacting drug pairs, stratified cross-validation becomes particularly valuable. This variant preserves the original class distribution within each fold, ensuring that the proportion of positive and negative samples remains consistent across all training and testing subsets. Maintaining class balance prevents biased performance estimates that could arise if some folds contain disproportionately few positive interaction examples, thereby enhancing the robustness and clinical relevance of the model evaluation. Together, these cross-validation strategies are essential for optimizing machine learning models in DDI prediction, facilitating reliable performance assessment even in the context of limited or imbalanced datasets (72).

7.3 External Validation and Benchmarking Datasets

To rigorously assess the generalizability of drug-drug interaction (DDI) predictive models, external validation using independent datasets is indispensable. Unlike cross-validation, which primarily evaluates model robustness within a single dataset, external validation tests the model’s performance on entirely separate and unseen data sources. This step is critical to confirm that predictive models maintain accuracy and reliability when applied beyond their original training environment, thereby supporting their practical utility in diverse clinical settings (73). Several widely accepted benchmark datasets serve as standard resources for external validation in DDI modeling. These include:

Drug Bank, which provides comprehensive, curated information on approved and experimental drugs, including detailed interaction profiles.

TWOSIDES, a database compiling statistically inferred adverse drug interaction side effects derived from large-scale clinical datasets.

SIDER, a resource cataloging marketed drugs alongside their documented adverse drug reactions, facilitating identification of clinically relevant DDI outcomes. Datasets curated from Bio Creative DDI extraction challenges, which contain annotated drug interaction data extracted from biomedical literature, supporting evaluation of text-mining based prediction methods. Utilizing these datasets for external validation offers several advantages. First, they establish a standardized framework for benchmarking, allowing researchers to objectively compare novel computational algorithms against established approaches. Second, testing models across multiple independent datasets reflects the heterogeneity and complexity of real-world data, including variations in drug combinations, patient populations, and clinical contexts. This broader evaluation enhances the confidence in model robustness and increases the likelihood of successful translation into clinical practice (74).

7.4 Interpretability and Explainability in DDI Models

As artificial intelligence (AI) models, particularly those based on deep learning and graph-based architectures, grow in complexity, ensuring interpretability and explainability has become a crucial requirement for fostering trust and promoting clinical adoption. Interpretability is defined as the degree to which human users can comprehend the underlying logic and decision-making process of a model, whereas explainability pertains to the ability to provide meaningful and understandable justifications for specific model predictions. To achieve these goals, various computational tools have been developed. Notably, SHAP (SHapley Additive explanations) and LIME (Local Interpretable Model-agnostic Explanations) are widely employed to elucidate the influence of individual features on a model’s output. These methods assign importance scores to input features, thereby allowing researchers and clinicians to assess whether the model’s decisions align with known biological mechanisms or pharmacological principles (75). In the context of drug-drug interaction (DDI) prediction, explainability techniques can reveal the molecular or mechanistic basis underlying a predicted interaction. This insight not only supports hypothesis generation for further experimental validation but also enhances drug safety evaluations by clarifying potential risk factors.Moreover, embedding explainable AI (XAI) practices within DDI predictive frameworks aligns with emerging ethical and regulatory standards in healthcare. Transparency in AI-driven decision-making processes is increasingly mandated to ensure patient safety, accountability, and fairness, ultimately facilitating broader acceptance and integration of AI-assisted drug interaction prediction tools in clinical practice (76).

8. APPLICATIONS AND USE CASES

The adoption of AI-based predictive modeling for drug-drug interactions (DDIs) has transitioned from purely academic research into practical applications across clinical, pharmaceutical, and regulatory domains. These models are proving instrumental in enhancing patient safety, streamlining drug development, and improving post-market surveillance.

8.1 Integration of Predictive Models into Clinical Decision Support Systems (CDSS)

One of the most transformative applications of artificial intelligence (AI)-driven drug-drug interaction (DDI) prediction lies in its incorporation within Clinical Decision Support Systems (CDSS). These systems are designed to assist healthcare professionals by providing evidence-based guidance during clinical workflows, particularly at the point of prescribing or medication reconciliation. By embedding AI-based DDI predictive models, CDSS can proactively identify and flag potentially harmful drug combinations, thereby significantly mitigating the risk of adverse drug events (ADEs) and improving patient safety outcomes. Conventional CDSS tools typically depend on static rule sets or curated interaction databases. While useful, these approaches are often limited by their inability to detect novel, rare, or complex DDIs that fall outside established knowledge bases. In contrast, AI-enhanced CDSS leverage dynamic algorithms capable of analyzing molecular structures, pharmacological mechanisms, and real-world clinical data. This enables a more comprehensive and context-sensitive evaluation of potential interactions, adapting to new evidence and diverse clinical scenarios. Moreover, advanced AI-powered CDSS have begun integrating patient-specific clinical parameters, such as age, renal function, hepatic status, and the presence of comorbidities, to tailor interaction alerts to individual risk profiles. Such personalized alerting systems help reduce the problem of “alert fatigue” — where clinicians become desensitized to frequent, non-specific warnings — thereby improving the clinical relevance and usability of interaction notifications. Hospitals employing these machine learning-driven DDI alerts have reported enhanced decision-making efficiency and a reduction in preventable medication errors, illustrating the practical benefits of AI integration into routine healthcare delivery (77).

8.2 Use in Drug Development and Post-Marketing Surveillance

Artificial intelligence (AI)-based drug-drug interaction (DDI) prediction is increasingly transforming various stages of drug discovery and development pipelines. During the preclinical and clinical phases, pharmaceutical companies utilize AI-driven predictive models to proactively identify potential interaction risks associated with new drug candidates. By anticipating harmful DDIs early in the development process, these models help mitigate the likelihood of costly late-stage failures, streamline the drug development timeline, and improve overall drug safety profiles before regulatory submission. In the post-marketing phase, AI models play a critical role in pharmacovigilance activities by continuously analyzing large-scale real-world data sources. These include electronic health records (EHRs), spontaneous adverse event reporting systems, and patient registries, which provide comprehensive and diverse patient-level information beyond the controlled environment of clinical trials. AI algorithms can detect emerging signals of previously unrecognized or rare DDIs, including those not captured during clinical studies due to limited sample sizes, population diversity, or study durations (78). Regulatory agencies such as the U.S. Food and Drug Administration (FDA) and other international counterparts have recognized the potential of AI technologies to enhance traditional pharmacovigilance methods. These bodies increasingly support the integration of AI tools to supplement existing safety monitoring frameworks, particularly for the early identification and evaluation of rare but clinically significant drug interactions, thereby contributing to improved public health outcomes (79).

8.3 Real-World Case Studies

Numerous real-world case studies have demonstrated the effectiveness of artificial intelligence (AI)-driven models in predicting drug-drug interactions (DDIs) and improving healthcare outcomes. These applications span drug discovery, clinical decision support, and pharmacovigilance, highlighting the broad utility of AI technologies in both research and clinical domains. One prominent example is IBM Watson for Drug Discovery, which integrates machine learning algorithms and natural language processing (NLP) techniques to mine extensive biomedical literature. This platform has been instrumental in uncovering previously unknown DDIs, particularly in complex therapeutic areas such as oncology and polypharmacy. By processing vast textual datasets, Watson has been able to provide actionable insights that inform drug safety research and guide clinical decisions in high-risk populations. In another case, researchers constructed a graph-based neural network model leveraging curated datasets from DrugBank and SIDER. This model was designed to predict novel DDIs by capturing the complex relationships between drugs, targets, and side effects within a structured network. The predictions were then validated using data from the FDA Adverse Event Reporting System (FAERS), and the model demonstrated superior performance in both precision and recall compared to traditional rule-based and machine learning approaches, reinforcing the potential of deep learning in DDI identification. A further example of clinical impact comes from a collaboration between Mayo Clinic and computational biology researchers, who implemented an AI-powered Clinical Decision Support System (CDSS) specifically tailored to identify harmful drug combinations frequently prescribed to elderly patients with multiple chronic conditions. By integrating patient-specific factors and pharmacological data, the system significantly reduced DDI-related hospitalizations, underscoring its role in enhancing patient safety, particularly in vulnerable populations. These case studies illustrate the transformative role of AI-based DDI prediction models not only in improving individual patient care but also in influencing broader public health strategies, regulatory frameworks, and comprehensive drug lifecycle management. As these technologies continue to evolve, they hold immense promise for advancing precision medicine and ensuring safer therapeutic practices (80).

9. CHALLENGES AND LIMITATIONS

While AI-based predictive modeling has significantly advanced the field of drug-drug interaction (DDI) prediction, several key challenges and limitations continue to hinder its full integration into clinical and pharmaceutical settings. These challenges range from data-related issues and model interpretability to regulatory barriers and ethical considerations.

9.1 Data Sparsity and Imbalance

A fundamental challenge in drug-drug interaction (DDI) predictive modeling is data sparsity, which arises because only a limited subset of all possible drug pairs has been experimentally tested or clinically documented. Considering the vast combinatorial space of potential drug combinations, the known and annotated DDIs represent only a small fraction, resulting in highly sparse interaction matrices that hinder comprehensive learning by AI models (81). In addition to sparsity, class imbalance poses a significant obstacle: the number of negative examples (i.e., drug pairs with no known interaction) greatly exceeds the number of positive, interacting pairs. This imbalance can bias predictive algorithms toward favoring the majority class, leading to an increased rate of false negatives and reducing the model’s sensitivity to true DDIs. These challenges of data sparsity and class imbalance not only limit the predictive accuracy but also impair the generalizability of models, especially when dealing with newly approved drugs or rare interaction events that lack sufficient training examples. To mitigate these issues, several strategies have been employed, including synthetic data generation through techniques like data augmentation, oversampling of minority classes, and cost-sensitive learning approaches that penalize misclassification of positive interactions more heavily. Despite these efforts, developing datasets that are both truly representative and balanced remains an ongoing challenge, underscoring the need for innovative solutions to enhance model robustness and reliability (82).

9.2 Black-Box Nature of Deep Models

Deep learning has become a cornerstone of modern drug-drug interaction (DDI) prediction, with numerous models based on architectures such as convolutional neural networks (CNNs), recurrent neural networks (RNNs), and graph neural networks (GNNs) showing promising predictive capabilities. However, a critical limitation of these models is their black-box nature—their internal decision-making processes are highly complex and not easily interpretable by human users. This opacity in reasoning poses substantial challenges, particularly in clinical contexts where decisions informed by AI must be transparent, explainable, and ethically justifiable. Physicians, pharmacists, and other healthcare professionals need to understand the basis of AI-generated predictions to confidently incorporate them into patient care, especially when those decisions involve potential risks to patient safety. To address these concerns, recent advancements in explainable artificial intelligence (XAI) have introduced methods aimed at improving interpretability of complex models. Tools such as Shapley Additive explanations (SHAP) and Local Interpretable Model-Agnostic Explanations (LIME) have been developed to attribute predictions to specific input features, providing insights into how individual variables contribute to the final output of an AI model. These tools offer post hoc explanations that are crucial for verifying the clinical plausibility of predicted DDIs and for fostering trust among medical practitioners. However, while XAI techniques have demonstrated potential, their applicability and reliability in high-stakes clinical environments remain an active area of research. Limitations include difficulty in interpreting explanations in very high-dimensional or interdependent data, and questions regarding the consistency and fidelity of the generated justifications. As such, further validation of these tools is necessary before they can be confidently relied upon in real-time clinical decision support systems (83).

9.3 Generalization to Unseen Drug Combinations

A significant and persistent limitation in artificial intelligence (AI)-based drug-drug interaction (DDI) prediction is the limited generalizability of models to previously unseen drug combinations. While many machine learning and deep learning models demonstrate high accuracy when evaluated on familiar compounds within established datasets, their performance often declines when applied to novel drugs, experimental agents, or drug repurposing scenarios. This limitation becomes particularly critical in contexts where rapid and accurate interaction assessments are needed—such as during global health emergencies (e.g., pandemics), or in personalized medicine, where patient-specific therapies may involve unique or non-standard drug combinations. The challenge arises partly due to the incomplete coverage of existing DDI datasets, which tend to be biased toward well-studied drugs and interactions. As a result, AI models trained on these datasets may overfit to known data distributions and struggle to extrapolate beyond them. This lack of extrapolative power restricts the clinical utility of AI systems in dynamic drug development pipelines and limits their effectiveness in emerging therapeutic areas.To address this issue, researchers have explored various strategies aimed at improving model generalizability. Transfer learning has been proposed as a promising approach, wherein knowledge gained from one domain or dataset is transferred to another related task involving novel drug entities. Data augmentation techniques, including the generation of synthetic drug pairs based on known interaction patterns, have also been employed to enrich training datasets and expose models to a broader chemical space. Additionally, incorporating chemical and biological similarity metrics—such as molecular fingerprints, target homology, or pathway overlap—into model architectures has shown potential for enhancing predictive accuracy on previously unseen drug combinations.  Despite these advancements, ensuring robust generalization across diverse drug spaces remains an open challenge in the field. Continued efforts are needed to design models that can reliably predict interactions involving newly approved drugs, off-label uses, or therapies introduced in response to emerging diseases (84).

9.4 Ethical, Legal, and Regulatory Concerns

While artificial intelligence (AI)-based models hold immense promise for enhancing drug-drug interaction (DDI) prediction, they also raise a number of ethical, legal, and regulatory concerns that must be carefully addressed to ensure their responsible and equitable use in healthcare settings. One of the foremost issues is data privacy, especially when using sensitive patient information from electronic health records (EHRs) for training predictive models. The utilization of such data must adhere to stringent privacy laws, including the Health Insurance Portability and Accountability Act (HIPAA) in the United States and the General Data Protection Regulation (GDPR) in the European Union, which mandate safeguards for patient confidentiality, informed consent, and data security (85). Another major concern is algorithmic bias, which can arise from imbalanced or non-representative training datasets. For instance, underrepresentation of certain demographic groups—such as racial minorities, older adults, or patients with rare diseases—can result in models that generate inequitable predictions, disproportionately affecting these populations and potentially reinforcing existing healthcare disparities. Addressing such biases requires deliberate efforts to ensure diversity in training data and transparency in model development processes. From a regulatory perspective, oversight of AI-driven DDI tools remains an evolving field. Regulatory agencies, including the U.S. Food and Drug Administration (FDA), have recognized the transformative potential of AI in drug safety monitoring and personalized medicine. However, these agencies emphasize the need for robust validation, clinical evidence, and post-market surveillance to ensure the safety, efficacy, and reliability of AI-based systems before and after deployment. Regulatory guidance documents increasingly call for transparency in model design, interpretability of outputs, and clearly defined accountability mechanisms, particularly when AI tools are integrated into clinical decision-making processes. Collectively, these ethical and regulatory dimensions underscore the importance of a responsible AI framework that promotes fairness, transparency, and accountability while safeguarding patient rights. Navigating these challenges is essential for building public trust and enabling the safe integration of AI technologies into clinical pharmacology and drug safety practices (86).

9.5 Integration into Clinical Practice

Despite substantial technical progress in the development of artificial intelligence (AI) models for drug-drug interaction (DDI) prediction, integration into clinical practice remains a considerable challenge. One of the primary obstacles is clinician skepticism regarding the reliability and usability of AI tools. Healthcare professionals may be reluctant to adopt these systems due to concerns about predictive accuracy, lack of interpretability, and the potential for workflow disruption during patient care. In high-stakes medical environments, tools that are not transparent or intuitive may be perceived as intrusive or even risky, especially if they generate false alerts or fail to provide justifiable recommendations. In addition, the technological infrastructure within many healthcare institutions is often inadequately prepared to support AI integration. Electronic health record (EHR) systems and clinical decision support systems (CDSS) may not be designed with the interoperability necessary to accommodate complex AI algorithms. The integration of AI models requires seamless data exchange, real-time processing, and compatibility with existing clinical workflows—requirements that are not universally met across health systems (87). Addressing these barriers will require more than just technical refinement of AI models. It demands a multifaceted implementation strategy involving interdisciplinary collaboration between data scientists, clinicians, informaticians, and administrators. Education and training programs should be developed to familiarize healthcare providers with the capabilities and limitations of AI-based tools, thereby enhancing user confidence and promoting responsible usage. Furthermore, clear operational guidelines must be established for the deployment, maintenance, and monitoring of these models in real-world healthcare environments, ensuring that AI integration enhances rather than disrupts clinical care. By focusing on these organizational and human factors—alongside technical innovation—stakeholders can facilitate successful adoption of AI-based DDI tools, ultimately contributing to safer prescribing practices and improved patient outcomes.

10. FUTURE DIRECTIONS

The evolution of AI-based predictive modeling for drug-drug interactions (DDIs) is poised to transform drug safety, clinical decision-making, and personalized therapy. Despite significant progress, future advancements must address current limitations and expand the scope of DDI prediction to make it more precise, interpretable, patient-centered, and ethically responsible. Several promising directions are emerging at the intersection of data science, medicine, and regulatory innovation.

10.1 Multimodal and Patient-Specific Data Integration

The next generation of drug-drug interaction (DDI) prediction models is anticipated to move beyond single-source data and embrace multimodal data integration as a core principle. This evolution involves the convergence of diverse and complementary data types—including genomics, proteomics, metabolomics, electronic health records (EHRs), drug labels, and biomedical literature—to develop more comprehensive and accurate representations of drug behavior and interaction mechanisms (88). By synthesizing these heterogeneous datasets, future models will be better equipped to capture the multifactorial and system-wide nature of drug interactions, which often vary based on biological context and population-specific characteristics. A major advancement will be the incorporation of patient-specific variables, such as genetic polymorphisms (e.g., variations in cytochrome P450 enzymes), comorbidities, hepatic or renal function, and concomitant therapies, all of which significantly influence pharmacokinetics and pharmacodynamics. This paradigm shift from population-level to individual-level modeling will enable context-aware DDI predictions, allowing for tailored assessments that consider the unique physiological and molecular profile of each patient. Such personalized modeling holds particular promise for improving drug safety and efficacy in vulnerable populations, including individuals experiencing polypharmacy, the elderly, or patients with rare diseases, where traditional DDI knowledge is often sparse or generalized. Additionally, advances in artificial intelligence (AI), such as deep learning and knowledge graph-based models, will further facilitate the dynamic integration of these multimodal inputs, enhancing predictive performance and clinical relevance. In summary, the future of AI-driven DDI prediction is characterized by a move toward precision pharmacology, where individualized risk assessments replace static, one-size-fits-all alerts, thus supporting safer, more effective, and equitable medication practices (89).

10.2 Personalized DDI Prediction and Precision Medicine

The incorporation of artificial intelligence (AI) into precision medicine is playing a transformative role in advancing personalized drug-drug interaction (DDI) prediction systems. Unlike traditional models that generalize interaction risks across broad populations, emerging AI frameworks are being designed to deliver individualized risk assessments by leveraging patient-specific data. These models integrate pharmacogenomic information, such as genetic polymorphisms influencing drug metabolism, along with longitudinal health records, including medication history, comorbidities, and clinical laboratory results, to more accurately predict how particular drug combinations may affect a specific patient (90). Such personalization enables dynamic risk stratification tailored to the patient’s current physiological and clinical context, allowing for proactive intervention. In addition to risk prediction, personalized DDI models contribute to dose optimization, suggesting adjusted dosing regimens based on predicted metabolic capacity or organ function. They also facilitate therapeutic substitution, recommending safer alternatives when high-risk interactions are identified—thus reducing the likelihood of adverse drug events (ADEs) and improving overall treatment outcomes. Looking forward, these intelligent systems could be further integrated into wearable health technologies, mobile health applications, or digital therapeutics platforms, offering real-time DDI alerts not only to clinicians but also to patients and caregivers. Such innovations would empower users with on-demand decision support, enhancing medication safety in outpatient and home-care settings where direct clinical supervision may be limited. This synergy between AI and precision medicine underscores the shift toward patient-centric pharmacotherapy, where treatment strategies are guided by individual data profiles, leading to more effective and safer healthcare delivery (91).

10.3 Explainable AI (XAI) in Pharmacovigilance

As artificial intelligence (AI) tools used in drug-drug interaction (DDI) prediction continue to evolve in complexity—particularly with the adoption of deep learning and graph-based neural networks—the need for explainable AI (XAI) has become increasingly urgent. Unlike traditional statistical models, which offer inherent transparency, many AI models operate as "black boxes", producing highly accurate outputs without providing insight into their internal reasoning. This lack of interpretability poses a significant barrier to clinical implementation, especially in high-stakes domains like pharmacovigilance, where decisions informed by AI can influence drug labeling, risk communication, or even regulatory approval and market withdrawal. To address this, future DDI models must prioritize transparent and interpretable outputs that are understandable and actionable by clinicians, researchers, and regulators alike. Explainability in this context goes beyond technical curiosity; it fosters trust, accountability, and regulatory compliance, ensuring that the AI system’s rationale aligns with known pharmacological mechanisms and clinical evidence. Several XAI techniques have emerged to enhance model interpretability. Notably, SHAP (SHapley Additive Explanations) provides a unified framework for attributing model predictions to specific input features, offering insights into how much each variable (e.g., enzyme inhibition, drug structure) contributed to a predicted DDI. Similarly, LIME (Local Interpretable Model-agnostic Explanations) approximates complex model behavior with locally interpretable surrogate models, revealing which factors most influenced a prediction in a particular case. Attention mechanisms, commonly used in transformer-based architectures, also help by highlighting relevant sections of molecular sequences or clinical data that the model “attends to” during prediction. Incorporating these XAI tools enables interpretation of DDI predictions in terms of chemical structure similarities, shared molecular pathways, or documented adverse clinical outcomes, making the models not only accurate but also scientifically and clinically meaningful. This approach is essential for integrating AI safely and effectively into drug development pipelines, regulatory workflows, and real-world clinical practice (92).

10.4 Federated Learning and Privacy-Preserving AI

Federated learning (FL) has emerged as a transformative paradigm in artificial intelligence (AI), particularly valuable in domains such as healthcare where data privacy and security are of paramount concern. Unlike conventional centralized learning frameworks, where data is aggregated into a central server for model training, FL allows AI models to be trained locally on decentralized data sources—such as individual hospitals, clinics, or healthcare institutions—without the need to transfer sensitive patient-level data. Instead of data, only model parameters or gradients are shared and aggregated centrally, thereby preserving privacy while still facilitating collaborative learning. This approach significantly enhances compliance with data protection regulations such as the Health Insurance Portability and Accountability Act (HIPAA) in the United States and the General Data Protection Regulation (GDPR) in the European Union. By ensuring that personally identifiable information (PII) remains within the originating institution, federated learning minimizes legal and ethical risks while maintaining the utility of real-world datasets for machine learning applications. In the context of drug-drug interaction (DDI) prediction, FL offers a promising solution to overcome the fragmentation of healthcare data across geographic and institutional boundaries. By enabling secure, multi-institutional collaboration, federated DDI models can benefit from heterogeneous patient populations, diverse prescribing patterns, and region-specific drug usage trends, thereby improving model robustness and generalizability. For example, data from hospitals treating geriatric patients in Europe can be combined with pediatric data from North America and oncology datasets from Asia, all without exposing patient identities. Moreover, federated frameworks can accelerate pharmacovigilance efforts, especially in detecting rare or delayed-onset DDIs that may not be evident in isolated datasets. The decentralized nature of FL also supports real-time model updates, enabling dynamic learning as new drug interactions or adverse events emerge in different parts of the world. By balancing data privacy, regulatory compliance, and AI-driven discovery, federated learning represents a crucial step forward in making large-scale, trustworthy, and globally relevant DDI prediction models a clinical reality (93).

10.5 Collaboration Among Academia, Healthcare, and Regulatory Bodies

The advancement and successful implementation of artificial intelligence (AI) models for drug-drug interaction (DDI) prediction rely fundamentally on interdisciplinary collaboration among various stakeholders. These stakeholders include researchers, clinicians, data scientists, pharmaceutical companies, and regulatory agencies, who must work together to establish and adhere to comprehensive standards for model validation, deployment, and ongoing monitoring to ensure safety, efficacy, and clinical utility. Such collaboration facilitates the development of robust, clinically relevant AI tools that are both scientifically sound and aligned with healthcare needs. For instance, researchers provide algorithmic innovation and technical expertise, clinicians contribute domain knowledge and practical insights, data scientists ensure proper handling and interpretation of complex datasets, pharmaceutical companies integrate models into drug development pipelines, and regulatory agencies oversee compliance with legal and ethical standards. Several collaborative initiatives exemplify this multidisciplinary approach. The U.S. Food and Drug Administration (FDA) has launched the Artificial Intelligence and Machine Learning (AI/ML) Action Plan, which outlines a framework for advancing regulatory science and fostering innovation in AI applications for healthcare. Similarly, the European Medicines Agency (EMA) has established the Big Data Task Force, aimed at creating guidance and policies for the use of big data and AI in medicine, including pharmacovigilance and drug safety assessment. Additionally, multi-institutional consortia such as Observational Health Data Sciences and Informatics (OHDSI) promote the harmonization of real-world data and open science principles to enhance collaborative research and benchmarking of AI models across diverse healthcare datasets .Together, these efforts demonstrate the critical importance of cooperation and shared governance in overcoming the technical, ethical, and regulatory challenges inherent in deploying AI-driven DDI prediction systems. They foster an ecosystem where innovation is balanced with patient safety and public trust, ultimately enabling the translation of AI advances into improved clinical outcomes. Such efforts are essential for aligning AI innovation with clinical needs and public health goals, ensuring that predictive DDI models are safe, equitable, and beneficial for all stakeholders (94).

CONCLUSION

Artificial intelligence has emerged as a transformative approach for predicting drug–drug interactions (DDIs), offering the capability to integrate heterogeneous biomedical data, model complex pharmacokinetic and pharmacodynamic relationships, and identify novel interaction patterns beyond the reach of conventional methods. By leveraging machine learning, deep learning, graph-based models, and natural language processing, AI-driven systems can analyze chemical structures, biological pathways, clinical records, and real-world pharmacovigilance data with remarkable scalability and precision. These models hold promise for early DDI detection, personalized risk assessment, and continuous post-marketing surveillance, thereby improving patient safety and optimizing therapeutic strategies. Despite these advances, several challenges remain. Data sparsity, imbalance, and variability across sources hinder model generalizability, while the “black-box” nature of many deep learning architectures raises concerns over interpretability and clinical trust. Ethical, regulatory, and infrastructural barriers particularly regarding data privacy, interoperability, and equitable performance across diverse populations must also be addressed before widespread clinical adoption is feasible. Future directions lie in multimodal and patient-specific data integration, explainable AI frameworks, privacy-preserving approaches such as federated learning, and stronger collaboration between academia, industry, and regulatory bodies. By aligning technological innovation with clinical needs and regulatory safeguards, AI-based predictive modeling can evolve into a reliable, transparent, and globally applicable tool for drug safety assessment. Ultimately, the convergence of AI, precision medicine, and pharmacovigilance has the potential to reshape how DDIs are predicted, prevented, and managed, fostering safer prescribing practices and more effective healthcare delivery.

REFERENCE

  1. Percha B, Altman RB. Informatics confronts drug–drug interactions. Trends in pharmacological sciences. 2013 Mar 1;34(3):178-84.
  2. Tatonetti NP, Ye PP, Daneshjou R, Altman RB. Data-driven prediction of drug effects and interactions. Science translational medicine. 2012 Mar 14;4(125):125ra31-.
  3. Li AP, Jurima-Romet M. Overview: pharmacokinetic drug-drug interactions. Advances in pharmacology. 1997 Jan 1; 43:1-6.
  4. Rombolà L, Scuteri D, Marilisa S, Watanabe C, Morrone LA, Bagetta G, Corasaniti MT. Pharmacokinetic interactions between herbal medicines and drugs: their mechanisms and clinical relevance. Life. 2020 Jul 4;10(7):106.
  5. Das S, Manjunath K, Biswas A, Sekar K. Therapeutic target identification and evaluation using machine learning. In Therapeutic Protein Targets for Drug Discovery and Clinical Evaluation: Bio-Crystallography and Drug Design 2023 (pp. 73-101).
  6. Sudsakorn S, Bahadduri P, Fretland J, Lu C. 2020 FDA drug-drug interaction guidance: a comparison analysis and action plan by pharmaceutical industrial scientists. Current drug metabolism. 2020 May 1;21(6):403-26.
  7. Hazell L, Shakir SA. Under-reporting of adverse drug reactions. Drug safety. 2006 May;29(5):385-96.
  8. Vilar S, Friedman C, Hripcsak G. Detection of drug–drug interactions through data mining studies using clinical sources, scientific literature and social media. Briefings in bioinformatics. 2018 Sep;19(5):863-77.
  9. Esteva A, Robicquet A, Ramsundar B, Kuleshov V, DePristo M, Chou K, Cui C, Corrado G, Thrun S, Dean J. A guide to deep learning in healthcare. Nature medicine. 2019 Jan;25(1):24-9.
  10. Ryu JY, Kim HU, Lee SY. Deep learning improves prediction of drug–drug and drug–food interactions. Proceedings of the national academy of sciences. 2018 May 1;115(18):E4304-11.
  11. Zhang W, Chen Y, Liu F, Luo F, Tian G, Li X. Predicting potential drug-drug interactions by integrating chemical, biological, phenotypic and network data. BMC bioinformatics. 2017 Jan 5;18(1):18.
  12. Neuvonen PJ, Niemi M, Backman JT. Drug interactions with lipid?lowering drugs: mechanisms and clinical relevance. Clinical Pharmacology & Therapeutics. 2006 Dec;80(6):565-81.
  13. Tanaka H, Mizojiri K. Drug-protein binding and blood-brain barrier permeability. The Journal of pharmacology and experimental therapeutics. 1999 Mar 1;288(3):912-8.
  14. Guengerich FP. Cytochrome p450 and chemical toxicology. Chemical research in toxicology. 2008 Jan 21;21(1):70-83.
  15. Levy G. Pharmacokinetics of salicylate elimination in man. Journal of pharmaceutical sciences. 1965 Jul 1;54(7):959-67.
  16. Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacology & therapeutics. 2013 Apr 1;138(1):103-41.
  17. Chen J, Raymond K. Roles of rifampicin in drug-drug interactions: underlying molecular mechanisms involving the nuclear pregnane X receptor. Annals of clinical microbiology and antimicrobials. 2006 Jan;5(1):3.
  18. Zhou S, Chan E, Lim LY, Boelsterli UA, Li SC, Wang J, Zhang Q, Huang M, Xu A. Therapeutic drugs that behave as mechanism-based inhibitors of cytochrome P450 3A4. Current drug metabolism. 2004 Oct 1;5(5):415-42.
  19. Rawlins MD, Thompson JW, Davies DM. Textbook of adverse drug reactions. by DM Davies. 1977:95.
  20. Katzung BG, Trevor AJ, Pacheco MD. Farmacologia. El Manual Moderno; 1991.
  21. Droogan C, Patel C, Yan GX, Kowey PR. Role of antiarrhythmic drugs: frequent implantable cardioverter-defibrillator shocks, risk of proarrhythmia, and new drug therapy. Heart Failure Clinics. 2011 Apr 1;7(2):195-205.
  22. Watanabe JH, McInnis T, Hirsch JD. Cost of prescription drug–related morbidity and mortality. Annals of Pharmacotherapy. 2018 Sep;52(9):829-37.
  23. Al-Ghazaly ZM, Jassim NA. Polypharmacy and potential drug–drug interactions in patients with rheumatoid arthritis. Medical Journal of Babylon. 2022 Jul 1;19(3):396-403.
  24. Trissel LA. Handbook on injectable drugs. InHandbook on Injectable Drugs 2007 (pp. 1720-1720).
  25. Newton DW. Drug incompatibility chemistry. American Journal of Health-System Pharmacy. 2009 Feb 15;66(4):348-57.
  26. Bourget P, Fernandez H. Incompatibilities and interactions of injectable drugs. Ann Pharmacother. 1999;33(3):347–354. doi:10.1345/aph.17217.
  27. Pirmohamed M, James S, Meakin S, et al. Adverse drug reactions as cause of admission to hospital: prospective analysis of 18,820 patients. BMJ. 2004;329(7456):15–19. doi:10.1136/bmj.329.7456.15.
  28. Budnitz DS, Lovegrove MC, Shehab N, Richards CL. Emergency hospitalizations for adverse drug events in older Americans. N Engl J Med. 2011;365(21):2002–2012. doi:10.1056/NEJMsa1103053.
  29. Nobili A, Garattini S, Mannucci PM. Multiple diseases and polypharmacy in the elderly: challenges for the internist of the third millennium. J Comorb. 2011; 1:28–44. doi:10.15256/joc.2011.1.13.
  30. Juurlink DN, Mamdani M, Kopp A, et al. Drug–drug interactions among elderly patients hospitalized for drug toxicity. JAMA. 2003;289(13):1652–1658. doi:10.1001/jama.289.13.1652.
  31. Wysowski DK, Nourjah P, Swartz L. Bleeding complications with warfarin use: a prevalent adverse effect resulting in regulatory action. Arch Intern Med. 2007;167(13):1414–1419. doi:10.1001/archinte.167.13.1414.
  32. Patel JP, Desai NR, Huang ES. Risk of hospitalization for bleeding with warfarin and NSAID co-administration. J Clin Pharm Ther. 2014;39(6):576–581. doi:10.1111/jcpt.12147.
  33. Bellosta S, Paoletti R, Corsini A. Safety of statins: focus on clinical pharmacokinetics and drug interactions. Circulation. 2004;109(23 Suppl 1): III50-57.
  34. Maher RL, Hanlon J, Hajjar ER. Clinical consequences of polypharmacy in elderly. Expert Opin Drug Saf. 2014;13(1):57–65. doi:10.1517/14740338.2013.827660.
  35. Mangoni AA, Jackson SHD. Age-related changes in pharmacokinetics and pharmacodynamics: basic principles and practical applications. Br J Clin Pharmacol. 2004;57(1):6–14. doi:10.1046/j.1365-2125.2003.02007. x.
  36. Spinewine A, Schmader KE, Barber N, et al. Appropriate prescribing in elderly people: how well can it be measured and optimized? Lancet. 2007;370(9582):173–184. doi:10.1016/S0140-6736(07)61091-0.
  37. European Medicines Agency. Guideline on the Investigation of Drug Interactions. EMA/CHMP/ICH/572198/2012; 2012.
  38. U.S. Food and Drug Administration. Guidance for Industry: Drug Interaction Studies — Study Design, Data Analysis, Implications for Dosing, and Labeling Recommendations. FDA; 2020.
  39. Flockhart DA. Drug Interactions: Cytochrome P450 Drug Interaction Table. Indiana University School of Medicine; 2007. Available from: https://drug-interactions.medicine.iu.edu/.
  40. Vilar S, Harpaz R, Uriarte E, Santana L, Rabadan R, Friedman C. Drug-drug interaction through molecular structure similarity analysis. J Am Med Inform Assoc. 2012;19(6):1066–1074. doi:10.1136/amiajnl-2012-000942.
  41. Ryu JY, Kim HU, Lee SY. Deep learning improves prediction of drug-drug and drug-food interactions. Proc Natl Acad Sci U S A. 2018;115(18):E4304–E4311. doi:10.1073/pnas.1803294115.
  42. FDA. Use of Computational Modeling and Simulation in Regulatory Review. Guidance for Industry. FDA; 2021.
  43. Lin JH, Lu AYH. Role of pharmacokinetics and metabolism in drug discovery and development. Pharmacol Rev. 1997;49(4):403–449.
  44. Houston JB. Utility of in vitro drug metabolism data in predicting in vivo metabolic clearance. Biochem Pharmacol. 1994;47(11):1823–1831.
  45. Rendic S, Di Carlo FJ. Human cytochrome P450 enzymes: a status report summarizing their reactions, substrates, inducers, and inhibitors. Drug Metab Rev. 1997;29(1-2):413–580.
  46. LeCluyse EL, Witek RP, Andersen ME, Powers MJ. Organotypic liver culture models: meeting current challenges in toxicity testing. Crit Rev Toxicol. 2012;42(6):501–548. doi:10.3109/10408444.2012.682115.
  47. Fahmi OA, Kharasch ED, Thummel KE. The cytochrome P450 3A inhibitor ketoconazole selectively inhibits intestinal CYP3A in humans. Clin Pharmacol Ther. 2009;85(6):573-579.
  48. Lin JH, Lu AYH. Role of pharmacokinetics and metabolism in drug discovery and development. Pharmacol Rev. 1997;49(4):403–449.
  49. Kuepfer L, Niederalt C, Wendl T, et al. Applied concepts in PBPK modeling: how to build a PBPK/PD model. CPT Pharmacometrics Syst Pharmacol. 2016;5(10):516–531. doi:10.1002/psp4.12110.
  50. Bailey DG, Dresser GK. Interactions between grapefruit juice and cardiovascular drugs. Am J Cardiovasc Drugs. 2004;4(5):281–297. doi:10.2165/00129784-200404050-00002.
  51. Temple R. Clinical drug interaction studies — study design, data analysis, and clinical implications. Clin Pharmacol Ther. 2010;87(6):718–720. doi:10.1038/clpt.2010.64.
  52. Vilar S, Harpaz R, Uriarte E, et al. Drug-drug interaction through molecular structure similarity analysis. J Am Med Inform Assoc. 2012;19(6):1066–1074. doi:10.1136/amiajnl-2012-000942.
  53. Tatonetti NP, Ye PP, Daneshjou R, Altman RB. Data-driven prediction of drug effects and interactions. Sci Transl Med. 2012;4(125):125ra31. doi:10.1126/scitranslmed.3003377.
  54. Preissner SC, Hoffmann MF, Preissner R. Computational approaches to predict drug-drug interactions: a review. Brief Bioinform. 2020;21(6):1802–1817. doi:10.1093/bib/bbz147.
  55. Zhang W, Chen Y, Liu F, et al. Predicting potential drug-drug interactions by integrating chemical, biological, phenotypic and network data. BMC Bioinformatics. 2017;18(1):18. doi:10.1186/s12859-016-1422-9.
  56. Topol EJ. High-performance medicine: the convergence of human and artificial intelligence. Nat Med. 2019;25(1):44–56. doi:10.1038/s41591-018-0300-7.
  57. Chen H, Engkvist O, Wang Y, Olivecrona M, Blaschke T. The rise of deep learning in drug discovery. Drug Discov Today. 2018;23(6):1241–1250. doi: 10.1016/j.drudis.2018.01.039.
  58. Zhang W, Chen Y, Liu F, et al. Predicting potential drug-drug interactions by integrating chemical, biological, phenotypic and network data. BMC Bioinformatics. 2017;18(1):18. doi:10.1186/s12859-016-1422-9.
  59. Beam AL, Kohane IS. Big data and machine learning in health care. JAMA. 2018;319(13):1317–1318. doi:10.1001/jama.2017.18391.
  60. Deo RC. Machine learning in medicine. Circulation. 2015;132(20):1920–1930. doi:10.1161/CIRCULATIONAHA.115.001593.
  61. Zhang W, Chen Y, Liu F, et al. Predicting potential drug-drug interactions by integrating chemical, biological, phenotypic and network data. BMC Bioinformatics. 2017;18(1):18. doi:10.1186/s12859-016-1422-9.
  62. Liu Y, Wei Q, Yu G, Gai L. An overview of computational methods for drug–drug interaction prediction. Curr Drug Metab. 2021;22(6):462–474. doi:10.2174/1389200222666210323122946.
  63. Rajkomar A, Dean J, Kohane I. Machine learning in medicine. N Engl J Med. 2019;380(14):1347–1358. doi:10.1056/NEJMra1814259.
  64. Chen H, Engkvist O, Wang Y, Olivecrona M, Blaschke T. The rise of deep learning in drug discovery. Drug Discov Today. 2018;23(6):1241–1250. doi: 10.1016/j.drudis.2018.01.039.
  65. Vamathevan J, Clark D, Czodrowski P, et al. Applications of machine learning in drug discovery and development. Nat Rev Drug Discov. 2019;18(6):463–477. doi:10.1038/s41573-019-0024-5.
  66. Liu Y, Wei Q, Yu G, Gai L. An overview of computational methods for drug–drug interaction prediction. Curr Drug Metab. 2021;22(6):462–474. doi:10.2174/1389200222666210323122946.
  67. Breiman L. Random forests. Mach Learn. 2001;45(1):5-32.
  68. Vilar S, Uriarte E, Santana L, et al. Detection of drug–drug interactions through data mining studies using clinical sources, scientific literature and social media. Brief Bioinform. 2018;19(4):693-710.
  69. Cortes C, Vapnik V. Support-vector networks. Mach Learn. 1995;20(3):273-297.
  70. Yamanishi Y, Kotera M, Kanehisa M, Goto S. Drug-target interaction prediction from chemical, genomic and pharmacological data in an integrated framework. Bioinformatics. 2010;26(12):i246-i254.
  71. Rogers D, Hahn M. Extended-connectivity fingerprints. J Chem Inf Model. 2010;50(5):742-754.
  72. Keiser MJ, Roth BL, Armbruster BN, et al. Relating protein pharmacology by ligand chemistry. Nat Biotechnol. 2007;25(2):197-206.
  73. Kuhn M, Letunic I, Jensen LJ, Bork P. The SIDER database of drugs and side effects. Nucleic Acids Res. 2016;44(D1):D1075-D1079.
  74. Ching T, Himmelstein DS, Beaulieu-Jones BK, et al. Opportunities and obstacles for deep learning in biology and medicine. J R Soc Interface. 2018;15(141):20170387.
  75. Goodfellow I, Bengio Y, Courville A. Deep Learning. MIT Press; 2016.
  76. Rifaioglu AS, Atas H, Martin MJ, et al. Recent applications of deep learning and machine intelligence on in silico drug discovery: methods, tools and databases. Brief Bioinform. 2020;21(6):1878-1912.
  77. Duvenaud DK, Maclaurin D, Aguilera-Iparraguirre J, et al. Convolutional networks on graphs for learning molecular fingerprints. Adv Neural Inf Process Syst. 2015; 28:2224-2232.
  78. Lee I, Keum J, Nam H. DeepConv-DTI: prediction of drug-target interactions via deep learning with convolution on protein sequences. PLoS Comput Biol. 2019;15(6): e1007129.
  79. Gilmer J, Schoenholz SS, Riley PF, et al. Neural message passing for quantum chemistry. Proc Mach Learn Res. 2017; 70:1263-1272.
  80. Hochreiter S, Schmidhuber J. Long short-term memory. Neural Comput. 1997;9(8):1735-1780.
  81. Zhang W, Liu F, Zhang C, et al. Predicting drug–drug interactions using a novel graph neural network with feature attention. BMC Bioinformatics. 2021;22(1):1-12.
  82. Wang F, Nie F, Huang H, et al. Drug–drug interaction extraction via convolutional neural networks. J Biomed Inform. 2017; 76:125-135.
  83. Lee H, Kang J, Hwang S, et al. Predicting drug–drug interactions using deep autoencoders. Front Pharmacol. 2020; 11:273.
  84. Wu Z, Pan S, Chen F, et al. A comprehensive survey on graph neural networks. IEEE Trans Neural Netw Learn Syst. 2021;32(1):4-24.
  85. Zitnik M, Agrawal M, Leskovec J. Modeling polypharmacy side effects with graph convolutional networks. Bioinformatics. 2018;34(13): i457-i466.
  86. Huang K, Xiao C, Glass LM, Sun J. MolRNN: molecular graph neural networks with recurrent neural networks for DDI prediction. J Chem Inf Model. 2021;61(2):647-659.
  87. Saito T, Rehmsmeier M. The precision-recall plot is more informative than the ROC plot when evaluating binary classifiers on imbalanced datasets. PLoS One. 2015;10(3):e0118432. doi: 10.1371/journal.pone.0118432.
  88. Chicco D, Jurman G. The advantages of the Matthews correlation coefficient (MCC) over F1 score and accuracy in binary classification evaluation. BMC Genomics. 2020;21(Suppl 1):6. doi:10.1186/s12864-019-6413-7.
  89. Arlot S, Celisse A. A survey of cross-validation procedures for model selection. Stat Surv. 2010; 4:40–79. doi:10.1214/09-SS054.
  90. Japkowicz N, Stephen S. The class imbalance problem: A systematic study. Intell Data Anal. 2002;6(5):429–49. doi:10.3233/IDA-2002-6504.
  91. Steyerberg EW, Harrell FE Jr. Prediction models need appropriate internal, internal-external, and external validation. J Clin Epidemiol. 2016; 69:245–7. doi: 10.1016/j.jclinepi.2015.04.005.
  92. Wishart DS, Feunang YD, Guo AC, Lo EJ, Marcu A, Grant JR, et al. DrugBank 5.0: A major update to the DrugBank database for 2018. Nucleic Acids Res. 2018;46(D1): D1074–82. doi:10.1093/nar/gkx1037.
  93. Tatonetti NP, Ye PP, Daneshjou R, Altman RB. Data-driven prediction of drug effects and interactions. Sci Transl Med. 2012;4(125):125ra31. doi:10.1126/scitranslmed.3003377.
  94. Kuhn M, Letunic I, Jensen LJ, Bork P. The SIDER database of drugs and side effects. Nucleic Acids Res. 2016;44(D1): D1075–9. doi:10.1093/nar/gkv1075.

Reference

  1. Percha B, Altman RB. Informatics confronts drug–drug interactions. Trends in pharmacological sciences. 2013 Mar 1;34(3):178-84.
  2. Tatonetti NP, Ye PP, Daneshjou R, Altman RB. Data-driven prediction of drug effects and interactions. Science translational medicine. 2012 Mar 14;4(125):125ra31-.
  3. Li AP, Jurima-Romet M. Overview: pharmacokinetic drug-drug interactions. Advances in pharmacology. 1997 Jan 1; 43:1-6.
  4. Rombolà L, Scuteri D, Marilisa S, Watanabe C, Morrone LA, Bagetta G, Corasaniti MT. Pharmacokinetic interactions between herbal medicines and drugs: their mechanisms and clinical relevance. Life. 2020 Jul 4;10(7):106.
  5. Das S, Manjunath K, Biswas A, Sekar K. Therapeutic target identification and evaluation using machine learning. In Therapeutic Protein Targets for Drug Discovery and Clinical Evaluation: Bio-Crystallography and Drug Design 2023 (pp. 73-101).
  6. Sudsakorn S, Bahadduri P, Fretland J, Lu C. 2020 FDA drug-drug interaction guidance: a comparison analysis and action plan by pharmaceutical industrial scientists. Current drug metabolism. 2020 May 1;21(6):403-26.
  7. Hazell L, Shakir SA. Under-reporting of adverse drug reactions. Drug safety. 2006 May;29(5):385-96.
  8. Vilar S, Friedman C, Hripcsak G. Detection of drug–drug interactions through data mining studies using clinical sources, scientific literature and social media. Briefings in bioinformatics. 2018 Sep;19(5):863-77.
  9. Esteva A, Robicquet A, Ramsundar B, Kuleshov V, DePristo M, Chou K, Cui C, Corrado G, Thrun S, Dean J. A guide to deep learning in healthcare. Nature medicine. 2019 Jan;25(1):24-9.
  10. Ryu JY, Kim HU, Lee SY. Deep learning improves prediction of drug–drug and drug–food interactions. Proceedings of the national academy of sciences. 2018 May 1;115(18):E4304-11.
  11. Zhang W, Chen Y, Liu F, Luo F, Tian G, Li X. Predicting potential drug-drug interactions by integrating chemical, biological, phenotypic and network data. BMC bioinformatics. 2017 Jan 5;18(1):18.
  12. Neuvonen PJ, Niemi M, Backman JT. Drug interactions with lipid?lowering drugs: mechanisms and clinical relevance. Clinical Pharmacology & Therapeutics. 2006 Dec;80(6):565-81.
  13. Tanaka H, Mizojiri K. Drug-protein binding and blood-brain barrier permeability. The Journal of pharmacology and experimental therapeutics. 1999 Mar 1;288(3):912-8.
  14. Guengerich FP. Cytochrome p450 and chemical toxicology. Chemical research in toxicology. 2008 Jan 21;21(1):70-83.
  15. Levy G. Pharmacokinetics of salicylate elimination in man. Journal of pharmaceutical sciences. 1965 Jul 1;54(7):959-67.
  16. Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacology & therapeutics. 2013 Apr 1;138(1):103-41.
  17. Chen J, Raymond K. Roles of rifampicin in drug-drug interactions: underlying molecular mechanisms involving the nuclear pregnane X receptor. Annals of clinical microbiology and antimicrobials. 2006 Jan;5(1):3.
  18. Zhou S, Chan E, Lim LY, Boelsterli UA, Li SC, Wang J, Zhang Q, Huang M, Xu A. Therapeutic drugs that behave as mechanism-based inhibitors of cytochrome P450 3A4. Current drug metabolism. 2004 Oct 1;5(5):415-42.
  19. Rawlins MD, Thompson JW, Davies DM. Textbook of adverse drug reactions. by DM Davies. 1977:95.
  20. Katzung BG, Trevor AJ, Pacheco MD. Farmacologia. El Manual Moderno; 1991.
  21. Droogan C, Patel C, Yan GX, Kowey PR. Role of antiarrhythmic drugs: frequent implantable cardioverter-defibrillator shocks, risk of proarrhythmia, and new drug therapy. Heart Failure Clinics. 2011 Apr 1;7(2):195-205.
  22. Watanabe JH, McInnis T, Hirsch JD. Cost of prescription drug–related morbidity and mortality. Annals of Pharmacotherapy. 2018 Sep;52(9):829-37.
  23. Al-Ghazaly ZM, Jassim NA. Polypharmacy and potential drug–drug interactions in patients with rheumatoid arthritis. Medical Journal of Babylon. 2022 Jul 1;19(3):396-403.
  24. Trissel LA. Handbook on injectable drugs. InHandbook on Injectable Drugs 2007 (pp. 1720-1720).
  25. Newton DW. Drug incompatibility chemistry. American Journal of Health-System Pharmacy. 2009 Feb 15;66(4):348-57.
  26. Bourget P, Fernandez H. Incompatibilities and interactions of injectable drugs. Ann Pharmacother. 1999;33(3):347–354. doi:10.1345/aph.17217.
  27. Pirmohamed M, James S, Meakin S, et al. Adverse drug reactions as cause of admission to hospital: prospective analysis of 18,820 patients. BMJ. 2004;329(7456):15–19. doi:10.1136/bmj.329.7456.15.
  28. Budnitz DS, Lovegrove MC, Shehab N, Richards CL. Emergency hospitalizations for adverse drug events in older Americans. N Engl J Med. 2011;365(21):2002–2012. doi:10.1056/NEJMsa1103053.
  29. Nobili A, Garattini S, Mannucci PM. Multiple diseases and polypharmacy in the elderly: challenges for the internist of the third millennium. J Comorb. 2011; 1:28–44. doi:10.15256/joc.2011.1.13.
  30. Juurlink DN, Mamdani M, Kopp A, et al. Drug–drug interactions among elderly patients hospitalized for drug toxicity. JAMA. 2003;289(13):1652–1658. doi:10.1001/jama.289.13.1652.
  31. Wysowski DK, Nourjah P, Swartz L. Bleeding complications with warfarin use: a prevalent adverse effect resulting in regulatory action. Arch Intern Med. 2007;167(13):1414–1419. doi:10.1001/archinte.167.13.1414.
  32. Patel JP, Desai NR, Huang ES. Risk of hospitalization for bleeding with warfarin and NSAID co-administration. J Clin Pharm Ther. 2014;39(6):576–581. doi:10.1111/jcpt.12147.
  33. Bellosta S, Paoletti R, Corsini A. Safety of statins: focus on clinical pharmacokinetics and drug interactions. Circulation. 2004;109(23 Suppl 1): III50-57.
  34. Maher RL, Hanlon J, Hajjar ER. Clinical consequences of polypharmacy in elderly. Expert Opin Drug Saf. 2014;13(1):57–65. doi:10.1517/14740338.2013.827660.
  35. Mangoni AA, Jackson SHD. Age-related changes in pharmacokinetics and pharmacodynamics: basic principles and practical applications. Br J Clin Pharmacol. 2004;57(1):6–14. doi:10.1046/j.1365-2125.2003.02007. x.
  36. Spinewine A, Schmader KE, Barber N, et al. Appropriate prescribing in elderly people: how well can it be measured and optimized? Lancet. 2007;370(9582):173–184. doi:10.1016/S0140-6736(07)61091-0.
  37. European Medicines Agency. Guideline on the Investigation of Drug Interactions. EMA/CHMP/ICH/572198/2012; 2012.
  38. U.S. Food and Drug Administration. Guidance for Industry: Drug Interaction Studies — Study Design, Data Analysis, Implications for Dosing, and Labeling Recommendations. FDA; 2020.
  39. Flockhart DA. Drug Interactions: Cytochrome P450 Drug Interaction Table. Indiana University School of Medicine; 2007. Available from: https://drug-interactions.medicine.iu.edu/.
  40. Vilar S, Harpaz R, Uriarte E, Santana L, Rabadan R, Friedman C. Drug-drug interaction through molecular structure similarity analysis. J Am Med Inform Assoc. 2012;19(6):1066–1074. doi:10.1136/amiajnl-2012-000942.
  41. Ryu JY, Kim HU, Lee SY. Deep learning improves prediction of drug-drug and drug-food interactions. Proc Natl Acad Sci U S A. 2018;115(18):E4304–E4311. doi:10.1073/pnas.1803294115.
  42. FDA. Use of Computational Modeling and Simulation in Regulatory Review. Guidance for Industry. FDA; 2021.
  43. Lin JH, Lu AYH. Role of pharmacokinetics and metabolism in drug discovery and development. Pharmacol Rev. 1997;49(4):403–449.
  44. Houston JB. Utility of in vitro drug metabolism data in predicting in vivo metabolic clearance. Biochem Pharmacol. 1994;47(11):1823–1831.
  45. Rendic S, Di Carlo FJ. Human cytochrome P450 enzymes: a status report summarizing their reactions, substrates, inducers, and inhibitors. Drug Metab Rev. 1997;29(1-2):413–580.
  46. LeCluyse EL, Witek RP, Andersen ME, Powers MJ. Organotypic liver culture models: meeting current challenges in toxicity testing. Crit Rev Toxicol. 2012;42(6):501–548. doi:10.3109/10408444.2012.682115.
  47. Fahmi OA, Kharasch ED, Thummel KE. The cytochrome P450 3A inhibitor ketoconazole selectively inhibits intestinal CYP3A in humans. Clin Pharmacol Ther. 2009;85(6):573-579.
  48. Lin JH, Lu AYH. Role of pharmacokinetics and metabolism in drug discovery and development. Pharmacol Rev. 1997;49(4):403–449.
  49. Kuepfer L, Niederalt C, Wendl T, et al. Applied concepts in PBPK modeling: how to build a PBPK/PD model. CPT Pharmacometrics Syst Pharmacol. 2016;5(10):516–531. doi:10.1002/psp4.12110.
  50. Bailey DG, Dresser GK. Interactions between grapefruit juice and cardiovascular drugs. Am J Cardiovasc Drugs. 2004;4(5):281–297. doi:10.2165/00129784-200404050-00002.
  51. Temple R. Clinical drug interaction studies — study design, data analysis, and clinical implications. Clin Pharmacol Ther. 2010;87(6):718–720. doi:10.1038/clpt.2010.64.
  52. Vilar S, Harpaz R, Uriarte E, et al. Drug-drug interaction through molecular structure similarity analysis. J Am Med Inform Assoc. 2012;19(6):1066–1074. doi:10.1136/amiajnl-2012-000942.
  53. Tatonetti NP, Ye PP, Daneshjou R, Altman RB. Data-driven prediction of drug effects and interactions. Sci Transl Med. 2012;4(125):125ra31. doi:10.1126/scitranslmed.3003377.
  54. Preissner SC, Hoffmann MF, Preissner R. Computational approaches to predict drug-drug interactions: a review. Brief Bioinform. 2020;21(6):1802–1817. doi:10.1093/bib/bbz147.
  55. Zhang W, Chen Y, Liu F, et al. Predicting potential drug-drug interactions by integrating chemical, biological, phenotypic and network data. BMC Bioinformatics. 2017;18(1):18. doi:10.1186/s12859-016-1422-9.
  56. Topol EJ. High-performance medicine: the convergence of human and artificial intelligence. Nat Med. 2019;25(1):44–56. doi:10.1038/s41591-018-0300-7.
  57. Chen H, Engkvist O, Wang Y, Olivecrona M, Blaschke T. The rise of deep learning in drug discovery. Drug Discov Today. 2018;23(6):1241–1250. doi: 10.1016/j.drudis.2018.01.039.
  58. Zhang W, Chen Y, Liu F, et al. Predicting potential drug-drug interactions by integrating chemical, biological, phenotypic and network data. BMC Bioinformatics. 2017;18(1):18. doi:10.1186/s12859-016-1422-9.
  59. Beam AL, Kohane IS. Big data and machine learning in health care. JAMA. 2018;319(13):1317–1318. doi:10.1001/jama.2017.18391.
  60. Deo RC. Machine learning in medicine. Circulation. 2015;132(20):1920–1930. doi:10.1161/CIRCULATIONAHA.115.001593.
  61. Zhang W, Chen Y, Liu F, et al. Predicting potential drug-drug interactions by integrating chemical, biological, phenotypic and network data. BMC Bioinformatics. 2017;18(1):18. doi:10.1186/s12859-016-1422-9.
  62. Liu Y, Wei Q, Yu G, Gai L. An overview of computational methods for drug–drug interaction prediction. Curr Drug Metab. 2021;22(6):462–474. doi:10.2174/1389200222666210323122946.
  63. Rajkomar A, Dean J, Kohane I. Machine learning in medicine. N Engl J Med. 2019;380(14):1347–1358. doi:10.1056/NEJMra1814259.
  64. Chen H, Engkvist O, Wang Y, Olivecrona M, Blaschke T. The rise of deep learning in drug discovery. Drug Discov Today. 2018;23(6):1241–1250. doi: 10.1016/j.drudis.2018.01.039.
  65. Vamathevan J, Clark D, Czodrowski P, et al. Applications of machine learning in drug discovery and development. Nat Rev Drug Discov. 2019;18(6):463–477. doi:10.1038/s41573-019-0024-5.
  66. Liu Y, Wei Q, Yu G, Gai L. An overview of computational methods for drug–drug interaction prediction. Curr Drug Metab. 2021;22(6):462–474. doi:10.2174/1389200222666210323122946.
  67. Breiman L. Random forests. Mach Learn. 2001;45(1):5-32.
  68. Vilar S, Uriarte E, Santana L, et al. Detection of drug–drug interactions through data mining studies using clinical sources, scientific literature and social media. Brief Bioinform. 2018;19(4):693-710.
  69. Cortes C, Vapnik V. Support-vector networks. Mach Learn. 1995;20(3):273-297.
  70. Yamanishi Y, Kotera M, Kanehisa M, Goto S. Drug-target interaction prediction from chemical, genomic and pharmacological data in an integrated framework. Bioinformatics. 2010;26(12):i246-i254.
  71. Rogers D, Hahn M. Extended-connectivity fingerprints. J Chem Inf Model. 2010;50(5):742-754.
  72. Keiser MJ, Roth BL, Armbruster BN, et al. Relating protein pharmacology by ligand chemistry. Nat Biotechnol. 2007;25(2):197-206.
  73. Kuhn M, Letunic I, Jensen LJ, Bork P. The SIDER database of drugs and side effects. Nucleic Acids Res. 2016;44(D1):D1075-D1079.
  74. Ching T, Himmelstein DS, Beaulieu-Jones BK, et al. Opportunities and obstacles for deep learning in biology and medicine. J R Soc Interface. 2018;15(141):20170387.
  75. Goodfellow I, Bengio Y, Courville A. Deep Learning. MIT Press; 2016.
  76. Rifaioglu AS, Atas H, Martin MJ, et al. Recent applications of deep learning and machine intelligence on in silico drug discovery: methods, tools and databases. Brief Bioinform. 2020;21(6):1878-1912.
  77. Duvenaud DK, Maclaurin D, Aguilera-Iparraguirre J, et al. Convolutional networks on graphs for learning molecular fingerprints. Adv Neural Inf Process Syst. 2015; 28:2224-2232.
  78. Lee I, Keum J, Nam H. DeepConv-DTI: prediction of drug-target interactions via deep learning with convolution on protein sequences. PLoS Comput Biol. 2019;15(6): e1007129.
  79. Gilmer J, Schoenholz SS, Riley PF, et al. Neural message passing for quantum chemistry. Proc Mach Learn Res. 2017; 70:1263-1272.
  80. Hochreiter S, Schmidhuber J. Long short-term memory. Neural Comput. 1997;9(8):1735-1780.
  81. Zhang W, Liu F, Zhang C, et al. Predicting drug–drug interactions using a novel graph neural network with feature attention. BMC Bioinformatics. 2021;22(1):1-12.
  82. Wang F, Nie F, Huang H, et al. Drug–drug interaction extraction via convolutional neural networks. J Biomed Inform. 2017; 76:125-135.
  83. Lee H, Kang J, Hwang S, et al. Predicting drug–drug interactions using deep autoencoders. Front Pharmacol. 2020; 11:273.
  84. Wu Z, Pan S, Chen F, et al. A comprehensive survey on graph neural networks. IEEE Trans Neural Netw Learn Syst. 2021;32(1):4-24.
  85. Zitnik M, Agrawal M, Leskovec J. Modeling polypharmacy side effects with graph convolutional networks. Bioinformatics. 2018;34(13): i457-i466.
  86. Huang K, Xiao C, Glass LM, Sun J. MolRNN: molecular graph neural networks with recurrent neural networks for DDI prediction. J Chem Inf Model. 2021;61(2):647-659.
  87. Saito T, Rehmsmeier M. The precision-recall plot is more informative than the ROC plot when evaluating binary classifiers on imbalanced datasets. PLoS One. 2015;10(3):e0118432. doi: 10.1371/journal.pone.0118432.
  88. Chicco D, Jurman G. The advantages of the Matthews correlation coefficient (MCC) over F1 score and accuracy in binary classification evaluation. BMC Genomics. 2020;21(Suppl 1):6. doi:10.1186/s12864-019-6413-7.
  89. Arlot S, Celisse A. A survey of cross-validation procedures for model selection. Stat Surv. 2010; 4:40–79. doi:10.1214/09-SS054.
  90. Japkowicz N, Stephen S. The class imbalance problem: A systematic study. Intell Data Anal. 2002;6(5):429–49. doi:10.3233/IDA-2002-6504.
  91. Steyerberg EW, Harrell FE Jr. Prediction models need appropriate internal, internal-external, and external validation. J Clin Epidemiol. 2016; 69:245–7. doi: 10.1016/j.jclinepi.2015.04.005.
  92. Wishart DS, Feunang YD, Guo AC, Lo EJ, Marcu A, Grant JR, et al. DrugBank 5.0: A major update to the DrugBank database for 2018. Nucleic Acids Res. 2018;46(D1): D1074–82. doi:10.1093/nar/gkx1037.
  93. Tatonetti NP, Ye PP, Daneshjou R, Altman RB. Data-driven prediction of drug effects and interactions. Sci Transl Med. 2012;4(125):125ra31. doi:10.1126/scitranslmed.3003377.
  94. Kuhn M, Letunic I, Jensen LJ, Bork P. The SIDER database of drugs and side effects. Nucleic Acids Res. 2016;44(D1): D1075–9. doi:10.1093/nar/gkv1075.

Photo
Sudarshan Gite
Corresponding author

B. Pharm, Gawande College of Pharmacy S. Kherda, Buldhana, Maharashtra, India.

Photo
Umesh Zanak
Co-author

B. Pharm, Gawande College of Pharmacy S. Kherda, Buldhana, Maharashtra, India.

Photo
Sakshi Bharate
Co-author

B. Pharm, Gawande College of Pharmacy S. Kherda, Buldhana, Maharashtra, India.

Photo
Pooja Rathod
Co-author

B. Pharm, Gawande College of Pharmacy S. Kherda, Buldhana, Maharashtra, India.

Photo
Vaishali Mawal
Co-author

B. Pharm, Gawande College of Pharmacy S. Kherda, Buldhana, Maharashtra, India.

Photo
Poonam Dalve
Co-author

B. Pharm, Gawande College of Pharmacy S. Kherda, Buldhana, Maharashtra, India.

Photo
Shivshankar Nagrik
Co-author

M. Pharm, Department of Pharmaceutics, Rajarshi Shahu College of Pharmacy Buldhana, Maharashtra, India

Sudarshan Gite*, Umesh Zanak, Sakshi Bharate, Pooja Rathod, Vaishali Mawal, Poonam Dalve, Shivshankar Nagrik, Artificial Intelligence in Predictive Modeling of Drug–Drug Interactions: Advances, Applications, and Future Directions, Int. J. Sci. R. Tech., 2025, 2 (8), 172-201. https://doi.org/10.5281/zenodo.16834759

More related articles
A Study on Partial Replacement of Cement with Rice...
Dr. Pranab Jyoti Barman, Abdul Masud, Anadi Krishna Saikia, Bhanu...
A Comprehensive Review on the Pharmacological Acti...
S. Swetha, M. Gopi, M. Gowri, V. Govarthini, K. Gowtham, ...
A Study on the Different Types of Biochar on the G...
Dr. Pranab Jyoti Barman, Abhisek Gupta, Debasish Dutta, Deepjyoti...
Olaparib: A Magic Bullet in the Treatment of Ovarian Cancer...
Bhagyashri Randhawan, Javane Nalini, Chaudhari Gauri, Damale Anjali, Munot Navinya, Mule Gayatri, Bh...
Chanting 'Om': A Neurochemical Pathway to Calm - Exploring GABAergic and Seroton...
Arnab Roy, Manish Kumar , Praveen Kumar Gupta , Mithun Kumar , Manish Kumar Singh , Gourav Nag, Adar...
Related Articles
The Interplay Between Calcium Homeostasis and Cardiac Function in Congestive Hea...
Arnab Roy, Mahesh Kumar Yadav, Naba Kishor Gorai , Rakhi Kumari , Megha Chattaraj , Tammana Parween,...
The Interplay Between Calcium Homeostasis and Cardiac Function in Congestive Hea...
Arnab Roy, Mahesh Kumar Yadav, Naba Kishor Gorai , Rakhi Kumari , Megha Chattaraj , Tammana Parween,...
A Comprehensive Review on Chikungunya Virus Epidemiology, Pathogenesis, Clinic...
Suraj Kedar, Prasad Harad, Bhavesh Mohape, Om Farde, Amin Shaikh, Manas Dandkar, Sarthak Bhusal, Nir...
The Effect of Size and Charge of Lipid Nanoparticles Prepared by Microfluidic Mi...
Sanchita Patil, Swaliha Mulla, Prajakta Mali, Sayali Shendage, Sakshi Kolekar, Deepak Kare, ...
A Study on Partial Replacement of Cement with Rice Husk Ash and Bamboo Biochar i...
Dr. Pranab Jyoti Barman, Abdul Masud, Anadi Krishna Saikia, Bhanujita Kutum, Chiranjib Gogoi, Madhur...
More related articles
A Study on Partial Replacement of Cement with Rice Husk Ash and Bamboo Biochar i...
Dr. Pranab Jyoti Barman, Abdul Masud, Anadi Krishna Saikia, Bhanujita Kutum, Chiranjib Gogoi, Madhur...
A Comprehensive Review on the Pharmacological Activities of Hylocereus Polyrhizu...
S. Swetha, M. Gopi, M. Gowri, V. Govarthini, K. Gowtham, ...
A Study on the Different Types of Biochar on the Geotechnical Behaviour of Local...
Dr. Pranab Jyoti Barman, Abhisek Gupta, Debasish Dutta, Deepjyoti Dutta, Jubli Jaman, Kangkana Gogoi...
A Study on Partial Replacement of Cement with Rice Husk Ash and Bamboo Biochar i...
Dr. Pranab Jyoti Barman, Abdul Masud, Anadi Krishna Saikia, Bhanujita Kutum, Chiranjib Gogoi, Madhur...
A Comprehensive Review on the Pharmacological Activities of Hylocereus Polyrhizu...
S. Swetha, M. Gopi, M. Gowri, V. Govarthini, K. Gowtham, ...
A Study on the Different Types of Biochar on the Geotechnical Behaviour of Local...
Dr. Pranab Jyoti Barman, Abhisek Gupta, Debasish Dutta, Deepjyoti Dutta, Jubli Jaman, Kangkana Gogoi...