View Article

Abstract

Drug-drug interactions (DDIs) represent a significant concern in healthcare, leading to adverse effects, altered drug efficacy, or therapeutic failure. Conventionally, DDIs are identified through clinical trials, in vitro experiments, or post-marketing surveillance, which are labor-intensive, time-consuming, and often insufficient to capture rare or complex interactions. The integration of artificial intelligence (AI) into drug development and clinical practice has introduced new possibilities in predicting potential DDIs, using vast datasets and advanced algorithms to foresee interactions before they occur. This review explores the key role of AI plays in identifying DDIs, the machine learning models applied, the importance of large databases in improving accuracy, and the challenges AI faces. We also discuss future directions for AI in creating more accurate, real-time DDI predictions, which could substantially reduce risks to patient safety and enhance personalized medicine.

Keywords

Artificial Intelligence, Drug-Drug Interactions, Machine Learning, Patient Safety, Predictive Modeling, Pharmacovigilance

Introduction

Drug-drug interactions are a leading cause of adverse drug reactions (ADRs) and can compromise treatment outcomes, especially in patients taking multiple medications. Polypharmacy, common among older adults and patients with chronic conditions, increases the risk of unintended DDIs (Krikorian G., 2021). Traditionally, the identification of DDIs has relied on retrospective studies, clinical trials, or case reports, often leaving gaps in our understanding of rare, complex, or context-dependent interactions (Chavda V.P., 2023) (Scannell J.W., 2012). The current approach may miss specific population subgroups or novel drug interactions, resulting in significant patient harm (B, 2009). AI offers a new approach to predict DDIs by leveraging machine learning and deep learning models that can process vast datasets, identify subtle patterns, and make accurate predictions that may not be immediately apparent to human researchers (Mak K.-K., 2019). AI systems can predict how different drugs interact based on their chemical structures, mechanisms of action, metabolic pathways, and real-world patient data. In this review, we examine how AI technology is being used to predict DDIs and its potential to revolutionize patient care by enabling healthcare professionals to avoid harmful interactions before they occur.

The Application of Ai In Predicting Drug-Drug Interactions:

Machine Learning Models for DDI Prediction: Machine learning (ML) models are at the forefront of AI applications in drug-drug interaction prediction. These models are trained on large-scale datasets, including drug properties, patient records, and known drug interactions, allowing them to predict potential DDIs based on learned patterns (Ashutosh Kumar Yadav, 2023). ML models can handle vast, multidimensional data, such as drug pharmacokinetics, genomics, and molecular structures, and can make predictions based on these complex relationships (JF, 1992). Several types of machine learning models are commonly employed in DDI prediction:

  • Support Vector Machines (SVM): SVMs are used to classify data by finding a hyperplane that separate interacting and non-interacting drug pairs based on their features (Sunarti S, 2021).
  • Random Forests: These ensemble learning methods build multiple decision trees to classify drug interactions, improving the model’s ability to handle large datasets and detect complex patterns (Agatonovic-Kustrin S, 2000).
  • Neural Networks: Deep learning neural networks, particularly known as convolutional neural networks (CNNs) or recurrent neural networks (RNNs) have been used to capture nonlinear relationships between drugs and their interaction profiles. They can also predict interactions in the absence of extensive human-curated data. Machine learning models offer significant advantages over traditional methods by continuously improving their prediction as more data is obtainable, resulting in increasingly accurate DDI predictions. Additionally, AI has the capability to predict interactions across multiple drug classes, which is particularly valuable in patients with polypharmacy (Zhang ZH, 2012).

Natural Language Processing (NLP) in Literature-Based DDI Prediction: Natural language processing (NLP) techniques have made significant advancements in automatically extracting potential drug interactions from sources like scientific literature, clinical guidelines, and adverse event reports. NLP algorithms are capable of processing vast quantities of unstructured data, such as research papers, case reports, and EHRs, to identify new or poorly understood drug interactions. AI-powered NLP systems can scan for terms related to DDIs, such as changes in drug metabolism, side effects, and contraindications, compiling this information into actionable insights (Ma J, 2015). By enabling the automated extraction and analysis of DDI-related data from diverse sources, NLP-driven AI systems can accelerate the identification of new or emerging drug interactions that may not yet be well-documented in clinical practice. This has proven to be especially useful for rare or unexpected DDIs which might only be appeared in isolated case reports or small-scale studies (M M. , 1995).

Data Sources for Ai-Driven Ddi Prediction:

Public Databases: The quality and accuracy of AI predictions largely depend on the datasets used to model training and validation. Several public databases serve as critical resources for DDI prediction by offering extensive datasets on drug interactions, pharmacological profiles, and adverse event reports:

  • Drug Bank: A comprehensive resource that integrates detailed chemical, pharmacological, and pharmaceutical data on drugs. Drug Bank offers a valuable information on drug interaction, including mechanisms of action, metabolism, and documented DDIs, making it an invaluable tool for AI-based DDI predictions (Shakya S, 2020).
  • FAERS (FDA Adverse Event Reporting System): This public database collects post-marketing adverse event data, including reports of drug interactions. AI models can use FAERS to identify potential DDIs based on real-world evidence from patient outcomes (J., 2017).
  • SIDER (Side Effect Resource): SIDER provides data on drug side effects and their frequencies. This resource allows AI systems model interactions based on shared adverse effects, which may indicate potential DDIs (Ramesh A.N, 2004).

Electronic Health Records (EHRs): EHRs are a rich source of real-world data, capturing detailed patient histories, treatment regimens, and clinical outcomes. By mining EHRs, AI models can identify patterns in how drugs interact in various patient populations, accounting for variables such as age, comorbidities, and concurrent medications. Additionally, EHR data can help refine DDI predictions by incorporating patient-specific factors, such as genetic predispositions and existing conditions that influence drug metabolism (Mandal L, 2019) (Hanson C.W, 2001).

The integration of EHR data into AI models allows for more personalized predictions of drug interactions, moving beyond generalized interactions to those specific to individual patients. This is particularly important in the context of polypharmacy, where the risk of DDIs is heightened, and individualized care is essential (Moore K.L, 2019) (Troulis M, 2002).

Ai Based Tools Used To Determine Drug- Drug Interaction:

  • IBM Watson for Drug Discovery:

This AI powered platform assists researchers to discover the potential drug-drug interactions by analyzing large datasets, including scientific literature and clinical trial data (IME, n.d.).

  • Medi-Span Clinical

An AI-driven platform that provides DDI screening and clinical decision support for healthcare professionals (Wolterskluwer, 2024).

  • DDInter (Deep Drug Interaction Predictor)

Uses deep learning algorithms to predict drug-drug interactions based on molecular structures and other pharmacological data.

  • DrugBank

An extensive AI-powered database that offers information about drug-drug interactions, along with drug targets, mechanisms, and more (DrugBank, n.d.).

  • Model-Informed Drug Development (MIDD)

 Leverages AI to predict drug-drug interactions (DDIs) early in the drug development process. This method combines clinical data, in-vitro studies, and AI to predict how drugs interact with enzymes and proteins, reducing adverse effects and increasing the success rate of clinical trials (VERISIMLife, 2024).

  • Caster

An AI framework, predicts DDIs by analyzing chemical substructures of drugs. It focuses on the relevant parts of drug molecules and provides interpretable results to healthcare professionals. This tool is designed to improve DDI prediction accuracy and manage adverse drug reactions more effectively (Kexin Huang, 2019).

The Role of Ai In Personalized Medicine:

Artificial intelligence is transforming personalized medicine, particularly in the context of predicting drug-drug interactions (DDIs) that tailored to individual patient profiles. Personalized medicine seeks to customize healthcare based on each patient such as genetics, lifestyle, and environment, relies heavily on the accurate prediction of DDIs (Arimura H, 2019). By combining real-world data with AI algorithms, personalized DDI predictions can offer more precise insights than generalized interactions, thereby improving therapeutic outcomes and reducing adverse reactions (Schmidt-Erfurth U, 2018) (Carreras J, 2022) (Poortmans P.M, 2020).In particular, AI models utilizing pharmacogenomics can predict how genetic variations in drug-metabolizing enzymes, such as CYP450, affect drug interactions in patients. For instance, a patient with a genetic variant that slows the metabolism of a specific drug may experience increased risks of interactions when that drug is combined with other medications. AI systems can integrate this data into predictive models, offering tailored dosing recommendations or alternative therapies based on individual genetic factors (Posner M.I, 2007) (Haag M, 2018).

AI and Polypharmacy: Polypharmacy, or the simultaneous use of multiple drugs, poses a significant challenge in clinical practice due to the heightened risk of DDIs. AI offers a solution by enabling healthcare providers to navigate the complexity of drug combinations more effectively. Through the application of machine learning algorithms, AI systems can analyze vast amounts of patient data, including drug regimens, medical histories, and clinical outcomes, to predict how combinations of medications will interact (M V. , 2018). AI-based tools are particularly useful in predicting interactions that involve not only two drugs but also complex regimens involving three or more medications, which are common in elderly populations or patients with multiple comorbidities. These models are continually refined as more data becomes available, making them an invaluable tool for managing polypharmacy and enhancing patient safety (Silver D, 2017) (Man KF, 2000).

Challenges and Limitations of Ai In Ddi Prediction:

While AI holds great promise in the prediction of DDIs, several challenges must be addressed to fully realize its potential in clinical settings. These challenges include data quality, model transparency, regulatory concerns, and the inherent complexity of drug interactions (Nikhil Singh, 2024).

Data Quality and Availability: AI models depend heavily on the availability and quality of data used to train them. Public databases, electronic health records (EHRs), and clinical studies provide vast amounts of information, but this data can be incomplete, biased, or outdated (Debleena Paulz, 2021). Furthermore, drug interaction data is often derived from post-marketing surveillance, which may not capture all potential interactions, especially rare or delayed reactions. Ensuring the completeness and accuracy of datasets is essential for developing robust AI systems capable of accurate DDI predictions. Efforts to integrate diverse data sources, such as real-world evidence, pharmacogenomic databases, and longitudinal patient records, will help improve AI models’ performance in predicting DDIs (Chai, 2020) (Thafar, 2019).

Model Interpretability and Transparency: Another significant limitation is the 'black box' nature of many machine learning models, particularly deep learning algorithms. Clinicians may hesitate to trust AI-driven DDI predictions if they cannot understand how the model arrived at a particular conclusion. Model interpretability is crucial for gaining clinician acceptance and for regulatory approval (Ozturk, 2018).

To overcome this challenge, efforts are being made to develop more transparent models, such as explainable AI (XAI), which provides insights into the decision-making process of AI systems. Such transparency could help bridge the gap between the sophisticated capabilities of AI and the practical needs of healthcare providers (Lounkine, 2012).

Future Directions of Ai In Ddi Prediction:

Integration of Multi-Omics Data: The future of AI-driven DDI prediction lies in the integration of multi-omics data, including genomics, proteomics, and metabolomics, to provide a more comprehensive view of how drugs interact within the human body. For example, genetic variations in drug-metabolizing enzymes like CYP450 could influence a patient’s risk of experiencing a DDI (Gao, 2018). This approach could allow for more precise predictions, leading to the development of personalized treatment regimens that minimize adverse interactions and improve therapeutic outcomes (Karimi, 2019). By combining pharmacogenomic data with machine learning models, future AI systems could predict DDIs at the individual patient level, significantly enhancing the safety and efficacy of drug therapies (Mayr, 2016).

Real-Time Monitoring Using AI: In the near future, AI could be integrated into electronic prescribing systems or even wearable devices that track drug administration in real-time. These systems would continuously monitor a patient’s medications and alert healthcare providers to potential DDIs before they become clinically significant. Real-time monitoring could also be enhanced through cloud-based platforms that use AI to compare ongoing treatments with current databases of known interactions, offering immediate insights to clinicians during patient care (Faure, 2001). Moreover, AI tools could be used for the development of mobile applications for patients, helping them understand possible DDIs when managing multiple medications. These tools would not only improve adherence to prescribed therapies but also empower patients to take proactive roles in their own healthcare (Das, 2016).

 

CONCLUSION:

Artificial intelligence represents a groundbreaking advancement in the prediction and management of drug-drug interactions, offering a faster, more efficient, and more precise approach than traditional methods. By leveraging large datasets, machine learning algorithms, and real-world evidence, AI-driven models can accurately predict both common and complex DDIs, potentially transforming clinical practice and enhancing patient safety. Despite the surrounding data quality, model transparency, and regulatory concerns, the integration of AI into healthcare holds enormous promise for the future of personalized medicine. Continued research and collaboration between AI developers, pharmacologists, and clinicians will be key to overcoming these obstacles and realizing the full potential of AI in DDI prediction.

REFERENCES

  1. Agatonovic-Kustrin S, B. R. (2000). Basic concepts of artificial neural network (ANN) modeling and its application in pharmaceutical research. J Pharm Biomed Anal, 717- 27.
  2. Arimura H, S. M. (2019). Radiomics with artificial intelligence for precision medicine inradiation therapy. J. Radiat. Res, 150-157.
  3. Ashutosh Kumar Yadav, A. Y. (2023). A review on drug stability. International Journal of Science and Research Archive, 474–485.
  4. B, M. (2009). Pichika M.R. Artificial Intelligence in Drug Development: Present Status and Future Prospects. Drug Discov. Today. , 959–968.
  5. Carreras J, N. N. (2022). Artificial IntelligenceAnalysis of Gene Expression Predicted the Overall Survival ofmantle Cell Lymphoma and a Large Pan-Cancer Series,. 155.
  6. Chai, S. (2020). A grand product design model for crystallization solvent design. Comput. Chem. Eng., 135.
  7. Chavda V.P., V. D. (2023). Bioinformatics Tools for Pharmaceutical Drug Product Development. Hoboken, NJ, USA: John Wiley & Sons, Ltd.
  8. Das, M. a. (2016). ANN in pharmaceutical product and process development. In Artificial Neural Network for Drug Design, Delivery and Disposition, 277-293.
  9. Debleena Paulz, G. S. (2021). Artificial intelligence in drug discovery and development. Drug Discovery Today, 80-92.
  10. DrugBank. (n.d.). Retrieved from AI-powered database : https://go.drugbank.com/
  11. Faure, A. e. (2001). Process control and scale-up of pharmaceutical wet granulation processes: a review. Eur. J. Pharm. Biopharm, 273- 277.
  12. Gao, K. e. (2018). Interpretable drug target prediction using deep neural representation. In Proceedings of the Twenty-Seventh International Joint Conference on Artificial Intelligence. IJCAI., 3371-3377.
  13. Haag M, M. L. (2018). Web-basedtraining. A new paradigm in computer-assisted instruction inmedicine,. J. Med. Inform, 72-76.
  14. Hanson C.W, M. B. (2001). Artificial intelligence applicationsin the intensive care unit. Crit Care Med, 427-235.
  15. IME. (n.d.). IBM Watson Health . Retrieved from IBM: https://www.ibm.com/search?lang=en&cc=us&q=IBM Watson Health
  16. J., M. (2017). On ‘Jeopardy’ Watson Win is All but Trivia. New York City: The New York Times.
  17. JF, D. (1992). Application of artificial intelligence to pharmacy and . 312-322.
  18. Karimi, M. e. (2019). DeepAffinity: interpretable deep learning of compound– protein affinity through unified recurrent and convolutional neural networks. Bioinformatics, 3329–3338.
  19. Kexin Huang, C. X. (2019, 11 15). MIT-IBM Watson AI Lab. Retrieved from CASTER: An AI framework for preventing adverse reactions to medication: https://mitibmwatsonailab.mit.edu/research/blog/caster-predicting-drug-interactions/
  20. Krikorian G., T. E. (2021). We Cannot Win the Access to Medicines Struggle Using the Same Thinking That Causes the Chronic Access Crisis. Health Hum. Rights, 119–127.
  21. Lounkine, E. e. (2012). Large-scale prediction and testing of drug activity on side-effect targets. Nature, 361-367.
  22. M, M. (1995). A comparison of classification in artificialintelligence. Induction versus a self-organising neural networks intelligence. Induction versus a self-organising neural networks. 117-128.
  23. M, V. (2018). Artificial intelligence. The beginning of a new era in pharmacy profession. Asian J. Pharm, 72-76.
  24. Ma J, S. R. (2015). Deep neural netsas a method for quantitative structure–activity relationships, . J Chem Inf Model, 263-274.
  25. Mak K.-K., P. M. (2019). Artificial Intelligence in Drug Development: Present Status and Future Prospects. Drug Discov. Today. , 773–780.
  26. Man KF, T. K. (2000). Genetic Algorithms, Concepts andDesigns, Assembly Automation,. 86-87.
  27. Mandal L, J. N. (2019). Prediction of Active Drug Molecule usingBack Propagation Neural Network. In Proceedings of the 8th. International Conference system Modeling and Advancement inResearch Trends, 22-26.
  28. Mayr, A. e. (2016). DeepTox: toxicity prediction using deep learning. . Frontiers Environ., 80.
  29. Moore K.L. (2019). Automated radiotherapy treatment planning. Philadelphia, PA, USA,: Seminars radiation Oncology, WB Saunder.
  30. Nikhil Singh, S. K. (2024). A Review on Artificial Intelligence in Pharma. International Journal of Pharmaceutical Sciences Review and Research, 33-39.
  31. Ozturk, H. e. (2018). DeepDTA: deep drug–target binding affinity prediction . Bioinformatics, i821-i829.
  32. Poortmans P.M, T. S. (2020). The use of Artificial Intelligence to individualiseradiation therapy for breast cancer, Breast. 194-200.
  33. Posner M.I, R. M. (2007). Research on Attention Networks asa Model for the Integration of Psychological Science, Psychol,. 1-23.
  34. Ramesh A.N, K. C. (2004). Artificialintelligence in medicine,. 334.
  35. Scannell J.W., B. A. (2012). Diagnosing the Decline in Pharmaceutical R&D Efficiency. Nat. Rev, 191–200.
  36. Schmidt-Erfurth U, S. A. (2018). Artificial intelligence in retina, Retin Eye Res,. 1-29.
  37. Shakya S. (2020). Analysis of artificial intelligence based imageclassification techniques. Journal of Innovative Image Processing, 44-54.
  38. Silver D, S. J. (2017). Mastering the game ofGo without human knowledge, . 354-359.
  39. Sunarti S, R. F. (2021). Artificial intelligence in healthcare Opportunities and Risk for future. S67–S70.
  40. Thafar, M. e. (2019). Comparison study of computational prediction tools for drug–target binding affinities. Frontiers Chem, 1-19.
  41. Troulis M, E. P. (2002). Developmentof a three-dimensional treatment planning system based oncomputed tomographic data,. 349-357.
  42. VERISIMLife. (2024, 4 1). Retrieved from Using AI & MIDD to Decode Drug-drug Interactions: https://www.verisimlife.com/publications-blog/using-ai-midd-to-decode-drug-drug-interactions
  43. Wolterskluwer. (2024). Retrieved from Medi-Span: Drug database solutions to strengthen healthcare decisions: https://www.wolterskluwer.com/en/solutions/medi-span
  44. Zhang ZH, W. Y. (2012). Development of glipizide push-pull osmotic pump controlled release tablets by using expert system and artificial neuranetwork. 1687-1695.

Reference

  1. Agatonovic-Kustrin S, B. R. (2000). Basic concepts of artificial neural network (ANN) modeling and its application in pharmaceutical research. J Pharm Biomed Anal, 717- 27.
  2. Arimura H, S. M. (2019). Radiomics with artificial intelligence for precision medicine inradiation therapy. J. Radiat. Res, 150-157.
  3. Ashutosh Kumar Yadav, A. Y. (2023). A review on drug stability. International Journal of Science and Research Archive, 474–485.
  4. B, M. (2009). Pichika M.R. Artificial Intelligence in Drug Development: Present Status and Future Prospects. Drug Discov. Today. , 959–968.
  5. Carreras J, N. N. (2022). Artificial IntelligenceAnalysis of Gene Expression Predicted the Overall Survival ofmantle Cell Lymphoma and a Large Pan-Cancer Series,. 155.
  6. Chai, S. (2020). A grand product design model for crystallization solvent design. Comput. Chem. Eng., 135.
  7. Chavda V.P., V. D. (2023). Bioinformatics Tools for Pharmaceutical Drug Product Development. Hoboken, NJ, USA: John Wiley & Sons, Ltd.
  8. Das, M. a. (2016). ANN in pharmaceutical product and process development. In Artificial Neural Network for Drug Design, Delivery and Disposition, 277-293.
  9. Debleena Paulz, G. S. (2021). Artificial intelligence in drug discovery and development. Drug Discovery Today, 80-92.
  10. DrugBank. (n.d.). Retrieved from AI-powered database : https://go.drugbank.com/
  11. Faure, A. e. (2001). Process control and scale-up of pharmaceutical wet granulation processes: a review. Eur. J. Pharm. Biopharm, 273- 277.
  12. Gao, K. e. (2018). Interpretable drug target prediction using deep neural representation. In Proceedings of the Twenty-Seventh International Joint Conference on Artificial Intelligence. IJCAI., 3371-3377.
  13. Haag M, M. L. (2018). Web-basedtraining. A new paradigm in computer-assisted instruction inmedicine,. J. Med. Inform, 72-76.
  14. Hanson C.W, M. B. (2001). Artificial intelligence applicationsin the intensive care unit. Crit Care Med, 427-235.
  15. IME. (n.d.). IBM Watson Health . Retrieved from IBM: https://www.ibm.com/search?lang=en&cc=us&q=IBM Watson Health
  16. J., M. (2017). On ‘Jeopardy’ Watson Win is All but Trivia. New York City: The New York Times.
  17. JF, D. (1992). Application of artificial intelligence to pharmacy and . 312-322.
  18. Karimi, M. e. (2019). DeepAffinity: interpretable deep learning of compound– protein affinity through unified recurrent and convolutional neural networks. Bioinformatics, 3329–3338.
  19. Kexin Huang, C. X. (2019, 11 15). MIT-IBM Watson AI Lab. Retrieved from CASTER: An AI framework for preventing adverse reactions to medication: https://mitibmwatsonailab.mit.edu/research/blog/caster-predicting-drug-interactions/
  20. Krikorian G., T. E. (2021). We Cannot Win the Access to Medicines Struggle Using the Same Thinking That Causes the Chronic Access Crisis. Health Hum. Rights, 119–127.
  21. Lounkine, E. e. (2012). Large-scale prediction and testing of drug activity on side-effect targets. Nature, 361-367.
  22. M, M. (1995). A comparison of classification in artificialintelligence. Induction versus a self-organising neural networks intelligence. Induction versus a self-organising neural networks. 117-128.
  23. M, V. (2018). Artificial intelligence. The beginning of a new era in pharmacy profession. Asian J. Pharm, 72-76.
  24. Ma J, S. R. (2015). Deep neural netsas a method for quantitative structure–activity relationships, . J Chem Inf Model, 263-274.
  25. Mak K.-K., P. M. (2019). Artificial Intelligence in Drug Development: Present Status and Future Prospects. Drug Discov. Today. , 773–780.
  26. Man KF, T. K. (2000). Genetic Algorithms, Concepts andDesigns, Assembly Automation,. 86-87.
  27. Mandal L, J. N. (2019). Prediction of Active Drug Molecule usingBack Propagation Neural Network. In Proceedings of the 8th. International Conference system Modeling and Advancement inResearch Trends, 22-26.
  28. Mayr, A. e. (2016). DeepTox: toxicity prediction using deep learning. . Frontiers Environ., 80.
  29. Moore K.L. (2019). Automated radiotherapy treatment planning. Philadelphia, PA, USA,: Seminars radiation Oncology, WB Saunder.
  30. Nikhil Singh, S. K. (2024). A Review on Artificial Intelligence in Pharma. International Journal of Pharmaceutical Sciences Review and Research, 33-39.
  31. Ozturk, H. e. (2018). DeepDTA: deep drug–target binding affinity prediction . Bioinformatics, i821-i829.
  32. Poortmans P.M, T. S. (2020). The use of Artificial Intelligence to individualiseradiation therapy for breast cancer, Breast. 194-200.
  33. Posner M.I, R. M. (2007). Research on Attention Networks asa Model for the Integration of Psychological Science, Psychol,. 1-23.
  34. Ramesh A.N, K. C. (2004). Artificialintelligence in medicine,. 334.
  35. Scannell J.W., B. A. (2012). Diagnosing the Decline in Pharmaceutical R&D Efficiency. Nat. Rev, 191–200.
  36. Schmidt-Erfurth U, S. A. (2018). Artificial intelligence in retina, Retin Eye Res,. 1-29.
  37. Shakya S. (2020). Analysis of artificial intelligence based imageclassification techniques. Journal of Innovative Image Processing, 44-54.
  38. Silver D, S. J. (2017). Mastering the game ofGo without human knowledge, . 354-359.
  39. Sunarti S, R. F. (2021). Artificial intelligence in healthcare Opportunities and Risk for future. S67–S70.
  40. Thafar, M. e. (2019). Comparison study of computational prediction tools for drug–target binding affinities. Frontiers Chem, 1-19.
  41. Troulis M, E. P. (2002). Developmentof a three-dimensional treatment planning system based oncomputed tomographic data,. 349-357.
  42. VERISIMLife. (2024, 4 1). Retrieved from Using AI & MIDD to Decode Drug-drug Interactions: https://www.verisimlife.com/publications-blog/using-ai-midd-to-decode-drug-drug-interactions
  43. Wolterskluwer. (2024). Retrieved from Medi-Span: Drug database solutions to strengthen healthcare decisions: https://www.wolterskluwer.com/en/solutions/medi-span
  44. Zhang ZH, W. Y. (2012). Development of glipizide push-pull osmotic pump controlled release tablets by using expert system and artificial neuranetwork. 1687-1695.

Photo
Abhishek Yadav
Corresponding author

Pharmacy College Azamgarh-276001(Uttar Pardesh)

Photo
Shweta Mishra
Co-author

Pharmacy College Azamgarh-276001(Uttar Pardesh)

Photo
Dr. Ashish Kumar Gupta
Co-author

Pharmacy College Azamgarh-276001(Uttar Pardesh)

Abhishek Yadav*, Shweta Mishra, Ashish Kumar Gupta, The Role of Artificial Intelligence in Predicting Drug-Drug Intraction: A New Frontier in Patient Safety, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 1, 2625-2631. https://doi.org/10.5281/zenodo.14780706

More related articles
Navigating Neurological Risks After COVID-19 Vacci...
Arnab Roy, K. Rajeswar Dutt, Ankita Singh, Mahesh Kumar Yadav, Ma...
A Detailed Review On "The Role Of Social Media In ...
Pratiksha Ganesh Ghodake, Shrushti Rahul Shahane, Utkarsha Ganesh...
Artificial Intelligence in Pharmacy: A Comprehensi...
Harshada Patil, Akshada Deshmukh, Komal Patil, Amruta Patil, Aish...
Related Articles
AI Powered Virtual Health Assistants: Transforming Patient Engagement Through Vi...
Durga P. Chavali, Vinod Kumar Dhiman, Siri Chandana Katari, ...
Effective Communication In Pharmacovigilance...
Prajakta Pujari , Sonali Magdum, Kajal Shinde, Darshani Taral, Prajakta Chaugule, Sadhana Pujari, Ra...
A Comprehensive Review On: Role Of Artificial Intelligence In Pharmaceutical Tec...
Yuthika Sudhir Ovhal, Ashwini Gavit, Kiran Makhija, N. S. Vyawhare, ...
Pharmacist’s Insight into Pharmacovigilance: An Exploratory Study...
LIJI C. J., Nafees K. Ismail, Ashbin Antony, Hemna Vinoj, ...
Navigating Neurological Risks After COVID-19 Vaccination Campaign: Insights And ...
Arnab Roy, K. Rajeswar Dutt, Ankita Singh, Mahesh Kumar Yadav, Manav Kumar, Kajal Kumari, Saurabh Ch...
More related articles
Navigating Neurological Risks After COVID-19 Vaccination Campaign: Insights And ...
Arnab Roy, K. Rajeswar Dutt, Ankita Singh, Mahesh Kumar Yadav, Manav Kumar, Kajal Kumari, Saurabh Ch...
A Detailed Review On "The Role Of Social Media In Modern Pharmacy: Impacts On Pr...
Pratiksha Ganesh Ghodake, Shrushti Rahul Shahane, Utkarsha Ganesh Sawant, Swapnil Kale, Sneha S. Kan...
Artificial Intelligence in Pharmacy: A Comprehensive Review of Applications, Dru...
Harshada Patil, Akshada Deshmukh, Komal Patil, Amruta Patil, Aishwarya Patil, Ishwari Nimbalkar, ...
Navigating Neurological Risks After COVID-19 Vaccination Campaign: Insights And ...
Arnab Roy, K. Rajeswar Dutt, Ankita Singh, Mahesh Kumar Yadav, Manav Kumar, Kajal Kumari, Saurabh Ch...
A Detailed Review On "The Role Of Social Media In Modern Pharmacy: Impacts On Pr...
Pratiksha Ganesh Ghodake, Shrushti Rahul Shahane, Utkarsha Ganesh Sawant, Swapnil Kale, Sneha S. Kan...
Artificial Intelligence in Pharmacy: A Comprehensive Review of Applications, Dru...
Harshada Patil, Akshada Deshmukh, Komal Patil, Amruta Patil, Aishwarya Patil, Ishwari Nimbalkar, ...