View Article

Abstract

Drug-drug interactions (DDIs) threaten patient safety and treatment results, especially with multiple drugs. Traditional methods like clinical trials and rule-based systems have limits in speed and scale. To improve detection, many now use artificial intelligence (AI) and machine learning (ML). These methods rely on a variety of data sources, including drug databases, patient records, and medical literature. They face challenges such as data quality, standardization, and missing information. Different AI models are used, like similarity-based systems, network models, graph neural networks, and deep learning approaches. Natural language processing (NLP) helps gather information from unstructured texts like clinical notes. AI tools are already used in hospitals and safety monitoring, guiding doctors and spotting rare interactions. Still, issues like model interpretation, data gaps, and fitting into clinical workflows remain. Future efforts focus on making AI more understandable, handling multiple data types, protecting privacy, and customizing medicine. Overall, AI is changing how DDIs are found and managed, helping improve drug safety and personalized treatment.

Keywords

Artificial Intelligence, Machine Learning, Deep Learning, Clinical Trials, Patient Safety, Pharmacovigilance.

Introduction

Background

Drug-drug interactions (DDI) occurs when one medicine changes how another works. This can make drugs less effective or cause new side effects. Some interactions are predictable, while others are not. They can be helpful, like when two medicines work well together, or harmful, causing problems. These interactions are important to watch out for to keep treatments safe and effective. These interactions can be classified into two main types namely, PK & PD (Figure 1)

Figure 1: Classification of Drug-drug Interactions

Pharmacokinetic Interactions:

  1. Absorption: Drug interactions can change how well a drug works. Sometimes, one drug affects how another drug is absorbed in the stomach or intestines E.g., antacids reducing absorption of some antibiotics.
  2. Distribution: Drugs can compete for binding sites on plasma proteins, affecting the distribution of each other.
  3. Metabolism: Drugs can induce or inhibit the enzymes responsible for metabolizing other drugs. E.g., rifampin speeds up metabolism of warfarin, reducing its effect.
  4. Excretion: The removal of drugs from the body, mainly via the kidneys.

E.g. probenecid inhibits the renal excretion of penicillin.

Pharmacodynamic Interactions:

  1. Additive Effects: When two drugs with similar pharmacological effects are taken together, their combined effect can be additive. For example, taking two antihypertensive drugs can lead to a greater reduction in blood pressure.
  2. Synergistic Effects: When the combined effect of two drugs is greater than the sum of their individual effects. For example, the combination of alcohol and benzodiazepines can lead to profound sedation.
  3. Antagonistic Effects: When one drug reduces or counteracts the effect of another drug. For example, naloxone antagonizes the effects of opioids.

Drug interactions cause many bad reactions, hospital stays, and higher healthcare costs. About 20-30% of bad drug events are due to these interactions. Serious issues like toxicity or failure of treatment can happen. Drugs like blood thinners, heart medicines, antibiotics, and mental health drugs are most likely to cause problems. Checking for interactions by hand relies on books or doctor knowledge, which can lead to mistakes. Computer tools that use fixed rules often show too many false alerts, making doctors ignore warnings. Static lists like Micromedex or Lexicomp may miss new interactions because they are not updated often enough.

Traditional Approaches to Drug-Drug Interaction (DDI) Prediction

  • Rule-Based Systems

Rule-based systems represent one of the earliest computational approaches utilized for drug-drug interaction (DDI) prediction. These systems are predicated on expert-defined rules that are based on pharmacokinetic (PK) and pharmacodynamic (PD) knowledge. Typically, they employ known drug properties, such as enzyme metabolism pathways (e.g., cytochrome P450), receptor binding affinities, and previously observed adverse effects, to identify potentially harmful interactions (Tatonetti et al., 2012). For instance, a rule may specify that if two drugs are substrates of the same enzyme, such as CYP3A4, their co-administration could lead to competitive inhibition, resulting in elevated plasma levels of one or both drugs. While rule-based systems are effective in identifying well-established interactions, they are inherently constrained by their dependence on existing knowledge bases. This limitation restricts their capacity to predict novel or complex interactions, particularly in polypharmacy scenarios or with drugs that have poorly understood mechanisms of action (Rasool et al., 2018).

  • Clinical trials and Post-Marketing Surveillance

Clinical trials test how safe and effective new drugs are, and sometimes they check for drug interactions. But early trials usually include only a small, similar group of people and are done in controlled settings. This makes it hard to see how drugs work in the real world with different patients. Ethical rules can also limit testing risky drug combos. So, trials might miss rare, long-term, or more complex interactions. After a drug is sold, the FDA relies on reports from doctors and patients to spot problems. These reports can find issues missed in trials but often have gaps. They may be incomplete or hard to link directly to the drug. So, post-marketing checks are useful but not perfect.

  • Limitations of Traditional Methods

Traditional methods for finding drug interactions are useful but have clear limits. Rule-based systems can't learn new interactions or adapt easily. Clinical trials are often too small, too slow, or too controlled to show all real-world issues. Watching drugs after they hit the market is helpful but takes time and can be biased. Because of these gaps, scientists are looking at artificial intelligence and machine learning. These new tools can analyse lots of data quickly, find hidden patterns, and predict possible interactions better than older methods. They offer more flexible and faster ways to detect drug-drug interactions, making medicine safer for everyone.

Enter AI: A New Paradigm

AI is changing how we predict drug-drug interactions. Traditional methods struggle with the large and varied data now available. AI handles complex information like drug structures, health records, side effects, and scientific papers. It finds hidden patterns better than old rule-based systems. AI models can analyze thousands of drug pairs quickly and can predict not just if drugs might interact, but how and how strongly. They can also use natural language processing to pull new knowledge from text sources and use relationships between drugs and proteins to see the bigger picture. AI can factor in individual patient details like age and genetics, making predictions more personal. It can simulate virtual patients to test drug combos first, saving time and money. Overall, AI helps catch problems early and finds new insights that can lead to safer, better treatments.

AI Techniques in Drug Interaction Prediction

  • Machine Learning Approaches

Drug–drug interactions (DDIs) pose a significant risk in clinical settings, especially given the growing number of patients on multiple medications. Traditional methods for DDI detection, such as clinical trials and in vitro/in vivo testing, are time-consuming, costly, and not scalable. To address these limitations, machine learning (ML) techniques have been widely adopted for both identifying known DDIs and predicting unknown interactions. This review outlines the primary datasets, methodologies, and challenges involved in ML-based DDI prediction, aiming to guide further development in this critical area of pharmacology.

  • Support Vector Machines (SVMs)

Support Vector Machines (SVMs) are supervised models used for classification, including drug-drug interaction (DDI) prediction. They work well with high-dimensional data and handle both simple and complex relationships. SVMs classify drug pairs as interacting or not, using features like structural similarity, side effects, gene expression, and protein data. They find a boundary that separates classes with the widest margin, which helps prevent overfitting on small datasets. By using kernel methods like RBF, SVMs can also learn non-linear patterns, making them effective for complex DDI tasks.

  • Random Forests                                                                                                                                                       

Random Forests are models that build many decision trees to make predictions. They are popular in drug interaction work because they handle different types of data well and don’t overfit easily. In drug-drug interaction prediction, they use info like drug structure, target similarity, and side effects. Each tree votes on whether two drugs might interact. The final answer is based on the most votes. Random Forests also tell us which features matter most. They work well even with noisy or incomplete data. They can find complex patterns without needing lots of tuning.

  • Gradient Boosting Machines (XGBoost, LightGBM)

Gradient Boosting Machines, like XGBoost and LightGBM, are strong models used for predicting drug-drug interactions (DDI). Unlike Random Forests, they build trees one after the other, fixing errors from previous trees, which helps improve accuracy. XGBoost is popular because it's fast, uses regularization, and works well with structured data. It is often used to analyze side effects, medication patterns, and molecular similarities. LightGBM is made by Microsoft and focuses on speed and saving memory. It's good for large datasets and can handle millions of drug pairs. Both models can use different goals, deal with missing data, and tell which factors matter most. Their success depends on tuning their settings and balancing the data, since most drug pairs don't interact.

  • DEEP LEARNING MACHINE
  • Artificial neural network (ANN)

Artificial neural networks (ANNs) are algorithms that find hidden patterns in data. They use many connected neurons to solve both simple and complex problems. Studies have used ANNs to predict drug-drug interactions (DDIs). Some models use two layers with different types of data, like similarity scores from various sources. Others use feed-forward networks with ReLU and sigmoid functions to process information. Some researchers also apply ANNs to graphs, where drugs are represented as nodes. They often combine ANNs with classifiers like XGBoost to determine if two drugs interact. These models help to improve the prediction of potential DDIs based on different data formats.

  • Convolutional neural network (CNN)

CNN is a type of neural network inspired by how animal visual parts work. It is good at handling grid-like data, such as images. The main goal of CNNs is to make data easier to process while keeping their ability to make accurate predictions. They do this with layers called convolution and pooling, which find specific features in small areas of the data. CNNs usually have three types of layers: convolution, pooling, and fully connected layers. The choice of activation function depends on the task. Sigmoid is often used for yes/no decisions, while softmax helps with multiple options. Several versions of CNNs have been created for predicting drug interactions, using different setups to improve accuracy.

  • Recurrent Neural Networks (RNNs)

Recurrent Neural Networks (RNNs) are a type of artificial brain that handles sequences of data. They can remember what came before and see how things are connected over time. This helps them spot patterns in how medicines are given to patients, especially when doses change or drugs are taken at different times. RNNs can learn how these patterns might cause side effects or interactions later. Some special versions, like LSTM or GRU, are better at keeping track of long-term info. They can also include details like dose size, how drugs are given, patient info, and lab results to improve accuracy. These tools help doctors understand how medicines work together over time, catching interactions that happen days or weeks later.

  • Transformers and Attention Mechanisms

Transformers changed how computers understand language. They use self-attention to consider all parts of a sentence at once. This helps them find relationships between words, even if they are far apart. The model was first shared in the paper “Attention is All You Need” in 2017. Since then, it has been used for medical and chemical texts. This helps find important drug information from unorganized writing. For predicting drug interactions, much useful info is in research papers, guidelines, drug labels, and reports. Older methods often missed details or misunderstood the context. Transformers are good at understanding the meaning behind words, making them better at detecting drug interactions hidden in text.

Language Models in Biomedical and Chemical Domains

Several transformer models have been pre-trained and fine-tuned specifically for biomedical and pharmacological text mining tasks:

BioBERT: -BioBERT is a language model for biomedical texts, built from BERT and trained on medical papers like PubMed abstracts. It improves tasks like finding medical names, understanding drug-disease relationships, and answering questions for drug interaction extraction. BioBERT identifies sentences showing drug effects, like "Drug A inhibits the metabolism of Drug B," and determines the interaction type. This helps understand drug interactions better.

DrugBERT: -DrugBERT is fine-tuned for drug-related tasks, including DDI extraction. Trained on annotated drug datasets, it excels at identifying drug mentions and relationships in medical texts. DrugBERT outperforms general models at handling pharmaceutical terminology.

ChemBERTa: ChemBERTa processes SMILES strings for chemical representation. It learns chemical features from molecular text to predict properties like toxicity and interactions. When combined with textual DDI extraction, it creates effective multi-modal DDI prediction systems.

  • Natural Language Processing (NLP)

Natural Language Processing (NLP) is crucial for predicting drug-drug interactions (DDIs) in biomedical AI. It processes unstructured texts like research papers and clinical notes, extracting key details into organized data to identify potential DDIs. NLP uses Named Entity Recognition (NER) to identify drug names and BioBERT models to understand context for interaction detection. Clinical notes provide real-world data, analyzed using ClinicalBERT. Tools like MetaMap and spaCy help identify drug mentions, working with knowledge graphs that map interactions. As technology advances, NLP continues improving drug safety through better detection of complex interactions.

Data Sources and Resources

The efficacy of artificial intelligence (AI) in drug-drug interaction (DDI) prediction is heavily contingent on the quality, variety, and comprehensiveness of the underlying data. A diverse range of data sources—spanning structured drug databases, clinical records, biomedical literature, and specialized DDI datasets—contribute to building robust predictive models. Each type of resource provides unique insights, from chemical structures and pharmacological mechanisms to real-world patient outcomes and scientific evidence. However, the heterogeneity and imperfections of these data sources present several preprocessing challenges that must be addressed to ensure model reliability. This section explores the key data resources used in AI-based DDI prediction, including drug databases, clinical data repositories, literature corpora, DDI-specific datasets, and the associated challenges in data preprocessing.

1. Drug Databases

Structured drug databases provide extensive information on drug properties, targets, and interactions. Key databases include DrugBank, PubChem, and ChEMBL.

DrugBank combines data on drugs, including chemicals, mechanisms, and targets. It contains information on FDA-approved drugs, biologics, supplements, and experimental medicines, along with reviewed drug interaction data from labels and trials. This makes it valuable for machine learning tools and knowledge graphs.

PubChem, by NCBI, is a public repository of chemical molecules and biological activities, providing compound structures and bioassay results. While not DDI-focused, it enables molecular-level analysis for DDI modeling through similarity-based inference.

ChEMBL, maintained by European Bioinformatics Institute, contains bioactive molecules with drug-like properties, providing binding, functional, and ADMET data. Its pharmacodynamic and pharmacokinetic profiles help train deep learning models for drug interaction understanding.

These databases provide structured features for machine learning and deep learning models in DDI prediction.

2. Clinical Data Sources

Clinical data provides insight into drug behavior across patient populations through EHRs, adverse event systems, and intensive care databases.

EHRs contain patient data including medication histories, diagnoses, and laboratory results. They help discover rare or population-specific DDIs and provide temporal information for studying sequential drug interactions. However, EHRs often contain inconsistencies requiring NLP techniques. FAERS collects adverse drug event reports from healthcare professionals, patients, and manufacturers. Mining FAERS reveals post-marketing DDI signals for validation.

MIMIC-III/IV comprises de-identified ICU patient data, including medications, vital signs, and clinical notes. It's used for DDI studies in critically ill patients and developing clinical AI tools. Integration of these clinical sources enables building context-aware models for patient-specific DDI factors.

3. Biomedical Literature

Scientific literature serves as a rich, unstructured resource containing DDI-related evidence from clinical trials, case reports, and pharmacological studies. PubMed and Medline are the two primary repositories used in this context.

  • PubMed, managed by the National Library of Medicine, indexes over 35 million citations for biomedical articles. Many DDIs are first reported in the literature, making PubMed a crucial resource for mining novel or emerging interactions. Researchers use NLP techniques such as named entity recognition (NER) and relation extraction to automate the identification of DDIs from abstracts and full texts.
  • Medline is the primary subset of PubMed, curated with controlled vocabulary indexing (MeSH terms). It facilitates more precise querying and semantic search capabilities. Literature-based data can be used to complement structured datasets and validate model outputs through cross-referencing with peer-reviewed findings.

Biomedical literature mining is essential for evidence-based AI models, enabling them to keep pace with the fast-evolving pharmacological knowledge landscape.

4. DDI-Specific Datasets

Specialized datasets focused on DDIs are critical for training and evaluating AI models. These datasets typically include labeled examples of interacting and non-interacting drug pairs, often with detailed annotations.

  • TWOSIDES is a large-scale dataset compiled from adverse event co-reporting in FAERS. It includes over 1,300 drug pairs and 1,300 side effects, facilitating the prediction of interaction-induced phenotypes. TWOSIDES is often used for evaluating models based on statistical inference, embeddings, and deep learning.
  • BioSNAP (Stanford Biomedical Network Analysis Project) provides curated biomedical interaction networks, including drug-drug interaction graphs. It includes both positive and negative samples and is commonly used to benchmark graph neural networks (GNNs) and link prediction algorithms.
  • SIDER (Side Effect Resource) contains information on marketed medicines and their recorded adverse effects. While not strictly a DDI dataset, SIDER can be combined with DDI corpora to understand the phenotypic consequences of interactions and improve model interpretability.

These datasets provide standardized benchmarks and allow the application of advanced AI techniques such as graph learning, multi-modal fusion, and ensemble modeling.

5. Data Preprocessing and Challenges

Despite abundant data, preprocessing remains a major bottleneck in developing reliable DDI prediction models. Key challenges include:

Missing Data: Clinical sources often contain incomplete records. Imputation techniques or models that handle missing values are needed.

Data Heterogeneity: Diverse formats from chemical structures to clinical notes require data integration pipelines. Standardization through ontologies enables harmonization.

Noise and Redundancy: FAERS and EHR sources can create spurious associations. Filtering, de-duplication, and noise-tolerant models are required.

Class Imbalance: DDI datasets have more negative than positive instances. SMOTE, cost-sensitive learning, and focal loss address this issue.

Annotation Inconsistency: Varying definitions in literature impact model generalization. Manual curation and dataset merging strategies help mitigate this.

Effective preprocessing is foundational to the accuracy and clinical relevance of DDI prediction systems.

Case Studies and Real-world Applications

The real-world integration of Artificial Intelligence (AI) in drug-drug interaction (DDI) prediction has significantly transformed pharmacology, enabling data-driven decisions, real-time alerts, and enhanced patient safety. This section provides a comprehensive examination of five landmark implementations of AI in DDI prediction and pharmacovigilance: DeepDDI, Decagon, transformer-based models for literature mining, Clinical Decision Support Systems (CDSS), and pharmacovigilance platforms such as IBM Watson and MedAware. These examples highlight the evolution of AI from theoretical frameworks to practical tools in healthcare.

  • DeepDDI: Leveraging Deep Learning on Drug Structural Information

DeepDDI is a deep learning tool that predicts how drugs might interact with each other. It analyzes the chemical structures of drugs using SMILES notation. The model has two main parts: one learns a math-based representation of each drug and the other uses these representations to guess the type of interaction. It can identify 86 different interaction types, like helpful effects or harmful reactions. DeepDDI performs better than older models and gives results that are easy to understand. This helps doctors and researchers make better decisions about drugs and safety.

  • Decagon: Graph Convolutional Networks for Multi-relational DDI Prediction

Decagon is a new method that uses graph convolutional networks to study drug interactions and side effects. Instead of looking at each drug pair alone, it creates a network of drugs and proteins. The network includes different types of links for interactions like binding or side effects. Decagon’s layers gather information from connected nodes to learn detailed features. It worked with data from over 900 drugs and nearly 1,000 side effects. The model predicted new side effects with high accuracy, many of which were later confirmed. This approach shows how combining data in a network can better understand complex drug interactions.

  • Transformer-based Models for Literature Mining

The rise of biomedical papers offers both chances and difficulties for predicting drug-drug interactions (DDIs). Extracting useful details from lots of unstructured text needs advanced natural language processing (NLP) tools. Transformer models like BERT and its versions, BioBERT and SciBERT, help with literature mining. These models are trained on large texts and fine-tuned for tasks such as recognizing drugs and finding interactions. They can identify drug names, categorize interaction types, and determine causality from scientific articles. Models trained on DDI data from sources like PubMed can automatically spot and classify interactions. They can link this information to standard systems like UMLS or MeSH to build structured databases. Compared to older methods, transformer models are more precise and find more interactions. They are now vital for updating DDI info quickly and learning from new findings in the field.

  • Integration into Clinical Decision Support Systems (CDSS)

AI tools that predict drug interactions are used in Clinical Decision Support Systems (CDSS) to help doctors make better choices. These systems analyze patient data, such as medications and health info, to spot problems and send alerts or advice in real time. This reduces side effects and helps doctors pick the right medicines. AI makes these alerts more accurate by filtering out irrelevant ones, lowering alert fatigue. Machine learning models trained on patient records find patterns of high-risk interactions and consider patient details like age and health conditions. Some systems use reinforcement learning to improve alerts based on doctor feedback, making them more user-friendly over time. Real-world use of these AI-enhanced systems shows they improve patient results, lower adverse drug events, and help follow clinical rules. They work especially well in intensive care units, where patients often take many drugs and quick action is vital.

  • AI in Pharmacovigilance Platforms: IBM Watson and MedAware

Pharmacovigilance—monitoring adverse effects of pharmaceutical products—has been enhanced by AI platforms IBM Watson for Drug Safety and MedAware.

IBM Watson uses natural language processing to extract safety signals from literature and clinical trials. It employs machine learning to identify adverse drug reactions and DDIs, supporting regulatory reporting and surveillance.

MedAware monitors EHRs using anomaly detection algorithms to flag prescription errors and DDIs in real time, delivering alerts for significant prescribing pattern deviations.

Both platforms demonstrate AI's power in detecting rare interactions, enhancing drug safety and compliance. Their deployment shows growing trust in AI within healthcare.

Challenges and Limitations

Despite the remarkable progress and widespread application of Artificial Intelligence (AI) in drug-drug interaction (DDI) prediction, several challenges and limitations continue to hinder its full potential and seamless integration into clinical practice. This section critically examines key barriers that include data quality and availability, the interpretability of AI models, generalization to unseen drugs, regulatory and ethical concerns, and difficulties in integrating AI systems with clinical workflows.

  • Data Quality and Availability

AI models for DDI prediction rely on data from EHRs, biomedical literature, reporting systems, and drug databases. However, these data sources vary significantly in quality and completeness. Datasets often contain missing entries, inconsistent nomenclature, and lack standardization across formats. This heterogeneity affects data integration and prediction accuracy. Clinical data, despite its value, remains largely inaccessible due to privacy regulations and proprietary constraints. Data imbalance presents another limitation, with known DDIs being overrepresented compared to non-interacting drug pairs. Rare or new drugs often lack sufficient historical data for training sets. Solutions require developing standardized, open-access DDI datasets and improved data augmentation techniques.

  • Interpretability of AI Models

AI models, especially deep learning ones, often work as "black boxes." They give predictions but don’t explain how they arrived at them. This is a problem in healthcare, where understanding results is key for trust. For example, models like DeepDDI and Decagon can predict harmful drug combos but don’t show why. Doctors may hesitate to act without knowing the reason behind a warning. People are trying ways to make these models more open, like using attention tools or methods like SHAP or LIME. Some are building models that are easier to understand on their own. But these methods often make the models less accurate or slow down their work. Many agree that medical AI should do both: be accurate and easy to explain. Achieving both goals is a tough challenge for research and real-world use.

  • Generalization to Unseen Drugs

Current AI models struggle to predict drug interactions for new drugs that weren’t in the training data. Since new drugs keep coming to the market, it’s hard for these models to guess how they will interact, especially if they are very different from known drugs. Deep learning models depend on patterns in their training data, so they often can’t handle unfamiliar cases well. Techniques like transfer learning and few-shot learning try to help by using what they already know about existing drugs to guess for new ones. These methods work best when the new drugs are similar to old ones. To do better, models need to keep learning as new data comes in. This means they must be updated regularly without losing the knowledge they already have. However, making these systems work this way is still a challenge.

  • Regulatory and Ethical Considerations

AI in healthcare must follow strict rules about data privacy, safety, and ethics. When predicting drug interactions, these rules cover patient consent, fairness, and responsibility. Agencies like the FDA and EMA are starting to give guidance on AI in medical devices, but clear rules for testing these AI tools are still rare. This makes it harder to use new models quickly. Ethical issues also come up. If AI learns from biased data, it can give unfair or wrong results, especially for some groups of people. If an AI suggests a bad drug combo, it’s unclear who is responsible. Is it the maker, the doctor, or the hospital? To fix these issues, developers should check for fairness and explain how their AI works. Regulators need to keep updating policies to keep people safe without stopping innovation.

  • Integration with Clinical Workflows

Advanced AI models must seamlessly integrate into clinical workflows to be valuable, as clinicians already face significant administrative burdens. A key challenge is AI system interoperability with health IT infrastructure, particularly EHRs. Data silos and incompatible formats hinder real-time access to information needed for AI predictions. Many facilities also lack resources to implement AI solutions. Clinicians must be trained to understand and use AI-generated outputs, requiring both technical training and cultural acceptance of AI as a collaborative tool. AI tools must prioritize usability and interoperability, with insights embedded in clinical dashboards and actionable recommendations to foster adoption.

Future Directions

AI is making great strides in predicting drug-drug interactions. In the future, we will see more use of explainable AI, which helps us understand how decisions are made. Combining data from different sources—called multimodal learning—can improve predictions. Privacy will be a big focus, with models that share information securely without exposing sensitive data. Predictions will connect more closely with personalized medicine, helping tailor treatments to individuals. Open science and collaboration platforms will grow, making data and tools more accessible. All these steps will change how we predict, understand, and use drug interaction info in clinics and research.

Explainable AI (XAI) for DDI Prediction

Explainable AI (XAI) is becoming important in healthcare, especially for predicting drug interactions. Deep learning models are often like black boxes, making it hard for doctors and regulators to see how they work. XAI tools such as SHAP, LIME, and attention visualization help show which details influence the predictions. These details can include drug structures, interaction pathways, or patient info. Using XAI builds trust with doctors and helps developers make better systems. As rules for AI get stricter, XAI will be key to making drug predictions safe and clear for everyday use.

Multimodal Learning: Integrating Genomics, EHR, and Chemical Data

Traditional drug interaction models use only one type of data, like chemical info or study results. But drugs act in complex ways influenced by genetics, body functions, and environment. Multimodal learning combines different data types such as genetic info, health records, chemical structures, and side effect reports. This helps models understand drug interactions better, especially those affecting specific genetic groups. Recent studies show that neural network models like transformers and graph neural networks work better with multiple data types. They can predict more accurately but face challenges like mixing different data, missing info, and smaller datasets. Despite this, multimodal learning has strong potential to improve drug interaction predictions.

Federated Learning and Privacy-Preserving Models

Data privacy is a big challenge when using sensitive medical data for AI research. Federated learning helps solve this by training models across many hospitals or institutions without sharing raw data. Each place trains a local model and sends only the results, like model updates, to a central server. This keeps patient info safe and respects rules like HIPAA and GDPR. It allows different groups, like hospitals and drug companies, to share data in a way that improves predictions for drug interactions and other health issues. Techniques like privacy filters make this process even safer. While federated learning is still new in healthcare, early projects show it works well for tracking drug safety and spotting problems without risking privacy. This approach opens up more opportunities for safer, better AI in medicine.

Integration with Personalized Medicine

Personalized medicine focuses on creating treatments that fit each person based on their unique traits like genes, lifestyle, and environment. Combining drug interaction (DDI) prediction with personalized medicine is a natural and important step forward. AI models can use specific data from a person's genes, body chemistry, and medical history to predict drug interactions that are unique to them, not just general ones. For example, some people process drugs differently because of enzyme differences, which can cause interactions that standard models might miss. Personalized DDI prediction can make taking medicine safer, especially for older people with many health issues and medications. Systems that use personalized AI predictions can change drug plans as patient data changes, making treatments more effective and less harmful. This approach fits with trends in precise dosing, using genetic information for drug plans, and creating personalized treatment plans, offering a more active and patient-focused way to care for people.

Open Science and Collaborative DDI Prediction Platforms

Open science is changing how research is done and shared. In DDI prediction, open-source tools, public datasets, and teamwork platforms are speeding up progress and openness. Projects like BioSNAP, TWOSIDES, and OpenFDA give researchers easy-to-use datasets for consistent results and comparisons. Open-source libraries like DeepChem, Hugging Face, and PyTorch Geometric help quickly create and share AI models. Platforms like ELIXIR, OpenTargets, and AI groups support large-scale, reliable DDI prediction by promoting shared standards and tools. Open science also supports ethical AI by allowing model reviews and data transparency. As more people from universities, companies, and regulators join these efforts, AI-driven DDI prediction will get faster, broader, and safer. These changes are not just technical but also shift how drug interactions are understood and used in healthcare.

CONCLUSION

AI is changing how we find and predict drug-drug interactions (DDIs). Traditional methods can't handle today's complex drug use, but AI can analyze large amounts of data to spot hidden patterns. It uses information from chemical properties, patient reports, genetics, and clinical notes to predict both known and new interactions. Technologies like machine learning and deep learning help with these tasks by turning data into safety alerts. AI is also helping improve drug safety through systems that give doctors personalized advice, making treatment safer. But there are challenges. Many AI models act as "black boxes" and are hard to explain. They also struggle with new drugs or rare interactions because of limited data. Integrating AI into healthcare systems is not easy either. The future looks promising, with hopes for better explanations (explainable AI), safer data sharing, and personalized medicine. AI that combines different types of data can give a clearer picture of DDIs. Better tools and teamwork are speeding up progress. Overall, AI is becoming a key part of making drug use safer and more tailored to individuals. It still needs work, but it will change how we predict, prevent, and handle drug interactions for good.

REFERENCES

  1. Zhang, W., Chen, Y., Liu, F., Luo, F., Tian, G., & Li, X. (2017). Predicting potential drug-drug interactions by integrating chemical, biological, phenotypic and network data. BMC Bioinformatics, 18(1), 18.
  2. Vamathevan, J., Clark, D., Czodrowski, P., et al. (2019). Applications of machine learning in drug discovery and development. Nature Reviews Drug Discovery, 18(6), 463–477.
  3. Ryu, J. Y., Kim, H. U., & Lee, S. Y. (2018). Deep learning improves prediction of drug–drug and drug–food interactions. PNAS, 115(18), E4304–E4311.
  4. Kastrin, A., Ferk, P., & Leskošek, B. (2021). Predicting potential drug-drug interactions on the basis of drug interaction profiles. PLoS ONE, 16(1), e0245535.
  5. Zeng, X., Zhu, S., Liu, X., Zhou, Y., Nussinov, R., & Cheng, F. (2020). deepDR: a network-based deep learning approach to in silico drug repositioning. Bioinformatics, 35(24), 5191–5198.
  6. Gottlieb, A., Stein, G. Y., Ruppin, E., & Sharan, R. (2012). Predicting novel drug indications with machine learning. Molecular Systems Biology, 7, 496.
  7. Vilar, S., Harpaz, R., Uriarte, E., Santana, L., Rabadan, R., & Friedman, C. (2012). Drug–drug interaction through molecular structure similarity analysis. Journal of the American Medical Informatics Association, 19(6), 1066–1074.
  8. Chen, X., Yan, C. C., Zhang, X., Zhang, X., Dai, F., Yin, J., & Zhang, Y. (2016). Drug–target interaction prediction: databases, web servers and computational models. Briefings in Bioinformatics, 17(4), 696–712.
  9. Ryu, J. Y., Kim, H. U., & Lee, S. Y. (2018). Deep learning improves prediction of drug–drug and drug–food interactions. PNAS, 115(18), E4304–E4311.
  10. Zhang, P., Wang, F., Hu, J., & Sorrentino, R. (2015). Label propagation prediction of drug-drug interactions based on clinical side effects. Scientific Reports, 5, 12339.
  11. Liu, Y., Wei, Q., Yu, H., & Huang, J. (2020). Deep learning-based DDI prediction with adversarial multi-task learning. Bioinformatics, 36(17), 4685–4694.
  12. Han, Y., Wang, L., Liu, W., & Song, D. (2022). Multi-modal deep learning for predicting adverse drug-drug interactions. Briefings in Bioinformatics, 23(1), bbab435.
  13. Zitnik, M., Agrawal, M., & Leskovec, J. (2018). Modeling polypharmacy side effects with graph convolutional networks. Bioinformatics, 34(13), i457–i466. [Decagon]
  14. Luo, Y., et al. (2017). A network integration approach for drug-target interaction prediction and computational drug repositioning from heterogeneous information. Nature Communications, 8, 573.
  15. Lim, S., Lee, J., & Yoon, S. (2021). Drug–drug interaction extraction from the literature using a neural attention model with syntactic dependency features. Bioinformatics, 37(14), 1991–1997.
  16. Sun, H., et al. (2021). Graph convolutional networks for biomedical knowledge graph completion: A comparative study. BMC Bioinformatics, 22(1), 568.
  17. Lee, J., Yoon, W., Kim, S., et al. (2020). BioBERT: a pre-trained biomedical language representation model for biomedical text mining. Bioinformatics, 36(4), 1234–1240.
  18. Chen, Q., Allot, A., & Lu, Z. (2021). LitCoin NLP Challenge: extracting drug–drug interactions from literature using transformer models. Nature Scientific Data, 8, 198.
  19. Zhang, X., et al. (2022). ClinicalBERT-DDI: A contextualized representation learning model for DDI prediction from EHRs. Journal of Biomedical Informatics, 128, 104057.
  20. Kuhn, M., et al. (2016). The SIDER database of drugs and side effects. Nucleic Acids Research, 44(D1), D1075–D1079.
  21. Wishart, D. S., et al. (2018). DrugBank 5.0: A major update to the DrugBank database for 2018. Nucleic Acids Research, 46(D1), D1074–D1082.
  22. Tatonetti, N. P., et al. (2012). Data-driven prediction of drug effects and interactions. Science Translational Medicine, 4(125), 125ra31.
  23. Jagannatha, A. N., & Yu, H. (2016). Bidirectional RNN for medical event detection in electronic health records. NAACL-HLT, 473–482.
  24. Wright, A., et al. (2012). Clinical decision support capabilities of commercially-available clinical information systems. Journal of the American Medical Informatics Association, 19(3), 463–469.
  25. Sultana, J., et al. (2013). Clinical decision support systems and their effect on prescribing outcomes: A systematic review. Drugs & Aging, 30(10), 755–768.
  26. Phansalkar, S., et al. (2013). Drug–drug interactions that should be non-interruptive in order to reduce alert fatigue in electronic health records. Journal of the American Medical Informatics Association, 20(3), 489–493.
  27. Zhang, P., et al. (2017). Multi-label learning for DDI prediction using DDI corpus. BMC Bioinformatics, 18(Suppl 14), 509.
  28. Zeng, X., et al. (2021). AI4DDI: An AI platform for drug-drug interaction prediction using multiple data sources. Briefings in Bioinformatics, 22(6), bbab211.
  29. Chen, X., Yan, C. C., Zhang, X., et al. (2016). Drug–target interaction prediction: databases, web servers and computational models. Briefings in Bioinformatics, 17(4), 696–712.
  30. Sarker, A., et al. (2015). Utilizing social media data for pharmacovigilance: A review. Drug Safety, 38(8), 773–785.

Reference

  1. Zhang, W., Chen, Y., Liu, F., Luo, F., Tian, G., & Li, X. (2017). Predicting potential drug-drug interactions by integrating chemical, biological, phenotypic and network data. BMC Bioinformatics, 18(1), 18.
  2. Vamathevan, J., Clark, D., Czodrowski, P., et al. (2019). Applications of machine learning in drug discovery and development. Nature Reviews Drug Discovery, 18(6), 463–477.
  3. Ryu, J. Y., Kim, H. U., & Lee, S. Y. (2018). Deep learning improves prediction of drug–drug and drug–food interactions. PNAS, 115(18), E4304–E4311.
  4. Kastrin, A., Ferk, P., & Leskošek, B. (2021). Predicting potential drug-drug interactions on the basis of drug interaction profiles. PLoS ONE, 16(1), e0245535.
  5. Zeng, X., Zhu, S., Liu, X., Zhou, Y., Nussinov, R., & Cheng, F. (2020). deepDR: a network-based deep learning approach to in silico drug repositioning. Bioinformatics, 35(24), 5191–5198.
  6. Gottlieb, A., Stein, G. Y., Ruppin, E., & Sharan, R. (2012). Predicting novel drug indications with machine learning. Molecular Systems Biology, 7, 496.
  7. Vilar, S., Harpaz, R., Uriarte, E., Santana, L., Rabadan, R., & Friedman, C. (2012). Drug–drug interaction through molecular structure similarity analysis. Journal of the American Medical Informatics Association, 19(6), 1066–1074.
  8. Chen, X., Yan, C. C., Zhang, X., Zhang, X., Dai, F., Yin, J., & Zhang, Y. (2016). Drug–target interaction prediction: databases, web servers and computational models. Briefings in Bioinformatics, 17(4), 696–712.
  9. Ryu, J. Y., Kim, H. U., & Lee, S. Y. (2018). Deep learning improves prediction of drug–drug and drug–food interactions. PNAS, 115(18), E4304–E4311.
  10. Zhang, P., Wang, F., Hu, J., & Sorrentino, R. (2015). Label propagation prediction of drug-drug interactions based on clinical side effects. Scientific Reports, 5, 12339.
  11. Liu, Y., Wei, Q., Yu, H., & Huang, J. (2020). Deep learning-based DDI prediction with adversarial multi-task learning. Bioinformatics, 36(17), 4685–4694.
  12. Han, Y., Wang, L., Liu, W., & Song, D. (2022). Multi-modal deep learning for predicting adverse drug-drug interactions. Briefings in Bioinformatics, 23(1), bbab435.
  13. Zitnik, M., Agrawal, M., & Leskovec, J. (2018). Modeling polypharmacy side effects with graph convolutional networks. Bioinformatics, 34(13), i457–i466. [Decagon]
  14. Luo, Y., et al. (2017). A network integration approach for drug-target interaction prediction and computational drug repositioning from heterogeneous information. Nature Communications, 8, 573.
  15. Lim, S., Lee, J., & Yoon, S. (2021). Drug–drug interaction extraction from the literature using a neural attention model with syntactic dependency features. Bioinformatics, 37(14), 1991–1997.
  16. Sun, H., et al. (2021). Graph convolutional networks for biomedical knowledge graph completion: A comparative study. BMC Bioinformatics, 22(1), 568.
  17. Lee, J., Yoon, W., Kim, S., et al. (2020). BioBERT: a pre-trained biomedical language representation model for biomedical text mining. Bioinformatics, 36(4), 1234–1240.
  18. Chen, Q., Allot, A., & Lu, Z. (2021). LitCoin NLP Challenge: extracting drug–drug interactions from literature using transformer models. Nature Scientific Data, 8, 198.
  19. Zhang, X., et al. (2022). ClinicalBERT-DDI: A contextualized representation learning model for DDI prediction from EHRs. Journal of Biomedical Informatics, 128, 104057.
  20. Kuhn, M., et al. (2016). The SIDER database of drugs and side effects. Nucleic Acids Research, 44(D1), D1075–D1079.
  21. Wishart, D. S., et al. (2018). DrugBank 5.0: A major update to the DrugBank database for 2018. Nucleic Acids Research, 46(D1), D1074–D1082.
  22. Tatonetti, N. P., et al. (2012). Data-driven prediction of drug effects and interactions. Science Translational Medicine, 4(125), 125ra31.
  23. Jagannatha, A. N., & Yu, H. (2016). Bidirectional RNN for medical event detection in electronic health records. NAACL-HLT, 473–482.
  24. Wright, A., et al. (2012). Clinical decision support capabilities of commercially-available clinical information systems. Journal of the American Medical Informatics Association, 19(3), 463–469.
  25. Sultana, J., et al. (2013). Clinical decision support systems and their effect on prescribing outcomes: A systematic review. Drugs & Aging, 30(10), 755–768.
  26. Phansalkar, S., et al. (2013). Drug–drug interactions that should be non-interruptive in order to reduce alert fatigue in electronic health records. Journal of the American Medical Informatics Association, 20(3), 489–493.
  27. Zhang, P., et al. (2017). Multi-label learning for DDI prediction using DDI corpus. BMC Bioinformatics, 18(Suppl 14), 509.
  28. Zeng, X., et al. (2021). AI4DDI: An AI platform for drug-drug interaction prediction using multiple data sources. Briefings in Bioinformatics, 22(6), bbab211.
  29. Chen, X., Yan, C. C., Zhang, X., et al. (2016). Drug–target interaction prediction: databases, web servers and computational models. Briefings in Bioinformatics, 17(4), 696–712.
  30. Sarker, A., et al. (2015). Utilizing social media data for pharmacovigilance: A review. Drug Safety, 38(8), 773–785. 

Photo
Shivendra Singh
Corresponding author

SIRT -Pharmacy ,SAGE UNIVERSITY ,BHOPAL (M.P)

Photo
Shikha Lodhi
Co-author

SIRT -Pharmacy ,SAGE UNIVERSITY ,BHOPAL (M.P)

Shivendra Singh*, Shikha Lodhi, Artificial Intelligence in Drug Interaction Prediction, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 5, 2460-2473. https://doi.org/10.5281/zenodo.15425567

More related articles
Review on Regulatory Affairs in the Era of Artific...
Sushant Shinde , Prachi Telavane, Shubham Prabhu, Mohit Jadhav , ...
Emerging Technologies: Application Of Artificial I...
Atharva Dumbre , Saurabh Dushing , Shejal Dongare , Sanjana Gadek...
Cardiovascular Care Evolved: A Review of Evidence-Based Advances...
Syed Afnaan Naaz, Shinde Akanksha, Upase Narsing, Wadekar Vishal, Yadav Anil, Dr. Ashok Giri, ...
A Review on: Pharmacogenomics with AI (Clinical Sematic Network )...
Amol V. Supekar, Tagare Chetan B., Girhe Akshay R., Tanpure Siddharth S., Zirpe Pandhori , ...
Review Artical On Pharmacovigilance...
Aher Shailendra , Gajre Tushar , Bharitkar Rushikesh , Pagar Pratik , Mahankale Pratik , ...
Related Articles
A Review Of Artificial Intelligence In Drug Discovery And Development ...
Somnath S. Davkhar, Devadhe Sanika S., Khedkar Namdev Bhagwan, ...
Artificial Intelligence in Pharmacovigilance...
Shubhangi Dukare, Priti Pokharkar, Ganesh Lamkhade, Aditi Pawar, Vinit Bankar, ...
Enhancing Patient Care With AI Chatbots And Virtual Assistants...
Asma Shaikh , Haritha CK, Arina Mullick, Varun Gadia, ...
Diagnosis And Treatment of Alzheimer’s Disease Using Artificial Intelligence ...
Nageswara Rao Yatam, Chakradhar Chikkala, Veeranjaneyulu Donepalli, Narayana Raju Padala, Bhaskarara...
Review on Regulatory Affairs in the Era of Artificial Intelligence: Roles, Needs...
Sushant Shinde , Prachi Telavane, Shubham Prabhu, Mohit Jadhav , ...
More related articles
Review on Regulatory Affairs in the Era of Artificial Intelligence: Roles, Needs...
Sushant Shinde , Prachi Telavane, Shubham Prabhu, Mohit Jadhav , ...
Emerging Technologies: Application Of Artificial Intelligence and Machine Learni...
Atharva Dumbre , Saurabh Dushing , Shejal Dongare , Sanjana Gadekar, Rina Gaikwad , S.D.Mankar , ...
Review on Regulatory Affairs in the Era of Artificial Intelligence: Roles, Needs...
Sushant Shinde , Prachi Telavane, Shubham Prabhu, Mohit Jadhav , ...
Emerging Technologies: Application Of Artificial Intelligence and Machine Learni...
Atharva Dumbre , Saurabh Dushing , Shejal Dongare , Sanjana Gadekar, Rina Gaikwad , S.D.Mankar , ...