Appearance
This report is written by MaltSci based on the latest literature and research findings
How does molecular modeling predict drug interactions?
Abstract
Molecular modeling has emerged as a crucial tool in drug discovery, significantly enhancing our understanding of drug interactions at the molecular level. As the pharmaceutical industry faces increasing pressure to streamline drug development processes, the application of computational techniques has become essential for predicting binding affinities, assessing stability, and evaluating potential off-target interactions. This report provides a comprehensive overview of molecular modeling techniques, including molecular docking, molecular dynamics simulations, and quantitative structure-activity relationship (QSAR) modeling, which facilitate rational drug design and optimization of pharmacological profiles. Additionally, the integration of molecular modeling with experimental data is discussed, emphasizing the importance of data validation and cross-verification. Despite the advancements, challenges related to the accuracy of predictive algorithms and the quality of structural data remain significant hurdles. The future of molecular modeling in drug discovery lies in the integration of advanced computational methods and artificial intelligence, which promise to enhance predictive capabilities and address the complexities of drug interactions. In conclusion, the understanding of molecular interactions through modeling is paramount for the development of effective and safe therapeutic agents, with ongoing advancements expected to shape the future of drug discovery and development.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 Overview of Molecular Modeling Techniques
- 2.1 Molecular Docking
- 2.2 Molecular Dynamics Simulations
- 2.3 Quantitative Structure-Activity Relationship (QSAR) Modeling
- 3 Applications in Drug Discovery
- 3.1 Rational Drug Design
- 3.2 Optimization of Pharmacological Profiles
- 3.3 Off-Target Interaction Prediction
- 4 Integration with Experimental Data
- 4.1 Data Validation and Cross-Verification
- 4.2 Case Studies of Successful Integration
- 5 Challenges and Limitations
- 5.1 Accuracy of Predictive Models
- 5.2 Structural Data Quality
- 5.3 Computational Resource Constraints
- 6 Future Perspectives
- 6.1 Advances in Computational Techniques
- 6.2 Role of Artificial Intelligence in Molecular Modeling
- 7 Conclusion
1 Introduction
Molecular modeling has become an essential component in the realm of drug discovery and development, significantly enhancing our understanding of drug interactions at the molecular level. The rapid advancement of computational techniques has facilitated the exploration of the intricate dynamics between drugs and their biological targets, thereby enabling researchers to predict binding affinities, assess stability, and evaluate potential off-target interactions. This progress is particularly pertinent in an era where the pharmaceutical industry faces increasing pressure to streamline drug development processes, reduce costs, and improve therapeutic efficacy.
The significance of molecular modeling in drug discovery cannot be overstated. It allows for rational drug design, where computational tools are employed to optimize pharmacological profiles while minimizing adverse effects. Techniques such as molecular docking, molecular dynamics simulations, and quantitative structure-activity relationship (QSAR) modeling have emerged as pivotal methodologies in this domain. These approaches not only provide insights into the binding mechanisms of drugs but also help in understanding the conformational changes that occur upon drug-target interactions. Recent studies have highlighted the role of molecular modeling in cardiovascular pharmacology, showcasing its utility in predicting drug interactions and assessing cardiotoxicity [1]. Furthermore, the integration of artificial intelligence (AI) and big data analytics into molecular modeling has opened new avenues for predicting complex interactions, making it a vital tool for rationalizing drug interactions with their pharmacological targets [1].
Despite the advancements, the current landscape of molecular modeling is not without its challenges. Issues related to the accuracy of predictive algorithms, the quality of structural data, and the computational resources required for extensive simulations remain significant hurdles. For instance, while traditional approaches have been utilized to predict drug-polymer miscibility, molecular modeling has demonstrated superior accuracy in estimating binding energies and interaction sites [2]. Additionally, the growing complexity of drug-drug interactions necessitates the development of more sophisticated models that can capture the nuances of molecular interactions [3][4].
This report is organized to provide a comprehensive overview of the mechanisms through which molecular modeling predicts drug interactions. Following this introduction, we will delve into the various molecular modeling techniques, including molecular docking, molecular dynamics simulations, and QSAR modeling, in Section 2. Section 3 will explore the applications of these techniques in drug discovery, emphasizing rational drug design, optimization of pharmacological profiles, and the prediction of off-target interactions. In Section 4, we will discuss the integration of molecular modeling with experimental data, highlighting the importance of data validation and cross-verification through case studies. Section 5 will address the challenges and limitations faced in the field, particularly focusing on the accuracy of predictive models, structural data quality, and computational constraints. Lastly, we will present future perspectives on the advances in computational techniques and the role of AI in enhancing molecular modeling capabilities in Section 6, before concluding our findings in Section 7.
In summary, the understanding of molecular interactions through modeling is paramount for the development of effective and safe therapeutic agents. As the field continues to evolve, the integration of advanced computational methods with experimental validation will undoubtedly play a crucial role in shaping the future of drug discovery and development.
2 Overview of Molecular Modeling Techniques
2.1 Molecular Docking
Molecular modeling encompasses a range of computational techniques that predict the interactions between drug molecules and their biological targets, primarily proteins. Among these techniques, molecular docking stands out as a pivotal method used extensively in drug discovery.
Molecular docking is an in silico structure-based approach that allows for the prediction of how small molecules, known as ligands, bind to specific receptor sites on target proteins. This method predicts the binding pose, strength, and binding affinity of the ligand-receptor interactions using various scoring functions. The primary goal of molecular docking is to simulate the binding of ligands to their respective targets to facilitate the identification of potential therapeutic candidates[5].
The process begins with the selection of a target protein, often based on its relevance to a particular disease or biological pathway. Following this, a library of small molecules is screened using docking software that employs search algorithms to explore the conformational space of the ligands within the binding site of the protein. The software evaluates the interactions between the ligand and the protein, calculating an affinity scoring function that ranks the potential binding poses based on their estimated free energy of binding, often expressed in kcal/mol[6].
Advancements in molecular docking techniques have enhanced their applicability and accuracy. Traditional docking methods primarily relied on rigid docking approaches, which assumed that both the ligand and the protein remain static during the interaction. However, this does not accurately reflect the dynamic nature of biomolecular interactions. Recent developments have incorporated molecular dynamics simulations alongside docking to account for the flexibility of proteins and ligands, thereby improving the predictions of binding modes and affinities[5].
Moreover, the integration of deep learning (DL) models into molecular docking has further transformed the field. These models leverage large datasets to enhance the accuracy of predictions while reducing computational costs. DL approaches aim to incorporate the flexibility of proteins into docking predictions, addressing the limitations of traditional rigid docking methods. Despite these advancements, challenges remain, particularly in generalizing predictions beyond training datasets and accurately predicting molecular properties such as stereochemistry and steric interactions[7].
Molecular docking is not only applicable in traditional drug discovery but also in the identification of molecular targets for nutraceuticals and dietary supplements. This approach can elucidate the mechanisms of action of bioactive compounds found in food sources, aiding in the development of disease-specific therapies[8].
In summary, molecular docking serves as a critical tool in predicting drug interactions by simulating the binding of ligands to target proteins. The continuous evolution of docking methodologies, including the integration of molecular dynamics and deep learning, underscores its significance in modern drug discovery and the development of therapeutic strategies. The accurate modeling of these interactions is essential for advancing our understanding of pharmacology and improving drug design processes.
2.2 Molecular Dynamics Simulations
Molecular modeling techniques, particularly molecular dynamics (MD) simulations, have become indispensable tools in predicting drug interactions and understanding the underlying mechanisms of these interactions. Molecular docking and MD simulations work synergistically to provide insights into the binding modes, affinities, and stability of protein-ligand systems, which are crucial for structure-based drug design and optimization.
Molecular docking is a computational method that predicts how a ligand molecule binds to a specific receptor. It estimates the binding pose, strength, and affinity of the molecules through various scoring functions. This method has seen significant advancements, allowing for the efficient screening of potential drug candidates and providing a foundation for further simulations and experimental validation[5].
On the other hand, molecular dynamics simulations allow for the modeling of conformational changes that are critical during the binding process. These simulations enable researchers to calculate thermodynamic quantities essential for estimating binding affinities, thus enhancing the accuracy of predictions in drug discovery. Recent developments in computing capabilities and methodologies have transitioned MD simulations from theoretical constructs to practical applications in drug development[9].
MD simulations offer detailed atomic-level insights into the interactions between drugs and their targets, facilitating the optimization of drug delivery systems and the design of effective drug carriers. For instance, MD simulations have been applied to study various drug delivery systems, such as functionalized carbon nanotubes and chitosan-based nanoparticles, highlighting their drug-loading capacities and stability[10]. Furthermore, these simulations can assess the solubility of drugs and their transport pathways across biological membranes, which is critical for predicting drug behavior in vivo[11].
The impact of MD simulations extends to elucidating the binding mechanisms of drugs to their targets. For example, studies utilizing Gaussian accelerated molecular dynamics (GaMD) have provided insights into the pathways of drug binding to receptors, allowing for the prediction of drug-receptor interactions at an atomistic level. This enhanced sampling technique significantly accelerates simulations, making it feasible to conduct routine drug binding studies using standard computing resources[12].
Moreover, MD simulations can capture the dynamical and energetic aspects of protein-ligand interactions, which are essential for understanding the structure-function relationships in drug design. These simulations have been successfully applied in various stages of drug discovery, including virtual screening and the investigation of drug resistance mechanisms due to mutations in target proteins[13].
In summary, molecular modeling, particularly through the use of molecular dynamics simulations, plays a crucial role in predicting drug interactions by providing a comprehensive understanding of the molecular mechanisms involved. The integration of computational methods with experimental data enhances the predictive power of these simulations, facilitating the design of novel therapeutics and improving the efficiency of drug discovery processes[14][15].
2.3 Quantitative Structure-Activity Relationship (QSAR) Modeling
Molecular modeling, particularly through Quantitative Structure-Activity Relationship (QSAR) modeling, plays a crucial role in predicting drug interactions by establishing statistical correlations between the chemical structure of compounds and their biological activities. QSAR models utilize various molecular descriptors that represent the inherent properties of chemical compounds, allowing researchers to predict how these compounds will interact with biological targets, such as proteins or enzymes.
QSAR modeling involves several methodologies, which can be categorized based on the dimensionality of the data used. Traditional QSAR models typically analyze 1D or 2D molecular descriptors, which focus on the chemical properties of the ligands. However, recent advancements have introduced multi-dimensional approaches that consider additional factors such as induced fit and solvation scenarios, enhancing the predictive power of these models [16].
The integration of machine learning techniques with QSAR modeling has further revolutionized this field. For instance, the combination of chemoinformatics and QSAR has enabled the development of models that can efficiently predict molecular activities and analyze large datasets. This synergy allows for a more comprehensive understanding of the relationships between molecular structures and their biological effects [17]. Specifically, machine learning algorithms, including support vector machines and random forests, are employed to refine the predictive capabilities of QSAR models, making them more robust and applicable to diverse chemical spaces [18].
Moreover, QSAR models can be enhanced by utilizing hybrid approaches that integrate chemical structure information with biological assay data. This integration helps in overcoming limitations associated with traditional QSAR models, such as their restricted predictive power due to limited datasets [19]. By leveraging in vitro screening data and toxicogenomics information, researchers can create hybrid models that improve the accuracy of toxicity predictions and broaden the applicability of QSAR analyses across different chemical classes [20].
The development of multi-target QSAR models represents another significant advancement in this field. These models can simultaneously predict activities against multiple biological targets, thereby streamlining the drug discovery process by allowing researchers to assess a compound's potential efficacy against various diseases or conditions in a single analytical framework [21].
In conclusion, QSAR modeling serves as a powerful tool in molecular modeling for predicting drug interactions by correlating chemical structures with biological activities. Through advancements in machine learning and hybrid modeling approaches, QSAR continues to evolve, providing researchers with enhanced capabilities to predict and analyze the interactions of potential drug candidates effectively. This ongoing evolution is crucial for accelerating drug discovery and optimizing lead compounds in the pharmaceutical industry.
3 Applications in Drug Discovery
3.1 Rational Drug Design
Molecular modeling serves as a critical component in the rational drug design process, facilitating the prediction of drug interactions through various computational techniques. The understanding of biomolecular interactions hinges on the structural and dynamic details of the interacting systems, allowing researchers to implement rational structure-based drug design methodologies effectively. This integration of computational modeling with experimental approaches has made drug development more cost-efficient and targeted [22].
In rational drug design, molecular modeling techniques such as pharmacophore modeling, molecular dynamics simulations, virtual screening, and molecular docking are employed to elucidate the activity of biomolecules and define molecular determinants for interaction with drug targets. Kinases, for instance, are extensively studied due to their pivotal role in cellular functions, and computational methods are vital for designing effective kinase inhibitors as anticancer drugs [23].
Recent advancements in molecular simulations, big data analysis, and artificial intelligence have further enhanced the ability to rationalize drug interactions with their pharmacological targets. These in silico techniques are now being applied to various fields, including cardiovascular pharmacology, where they are used to assess drug interactions and cardiotoxicity [1]. The predictive capabilities of molecular dynamics simulations allow for modeling conformational changes critical to the binding process, which is essential for calculating thermodynamic quantities related to binding affinities [9].
Furthermore, the development of user-friendly computational tools has enabled medicinal chemists to utilize molecular modeling more frequently. For example, tools like VirtualDesignLab and MD Client assist in estimating binding affinities and exploring the stability of ligand-protein complexes, thereby aiding in the identification of potential improvements in drug candidates [24]. This reflects a shift towards a more integrated approach in drug discovery, where computational insights are combined with experimental validation to optimize drug design.
The applications of molecular modeling extend beyond predicting interactions; they also include the identification of novel drug targets and small molecular candidates through computer-aided drug design (CADD) techniques. These approaches leverage mathematical modeling to enhance the efficiency and accuracy of drug discovery, addressing the limitations of traditional methods [25]. Overall, molecular modeling not only streamlines the drug design process but also plays a pivotal role in understanding the complex interactions between drugs and their biological targets, ultimately contributing to the development of safer and more effective therapeutics.
3.2 Optimization of Pharmacological Profiles
Molecular modeling plays a pivotal role in predicting drug interactions, particularly in the context of drug discovery and optimization of pharmacological profiles. The methodologies employed in molecular modeling allow researchers to elucidate the intricate details of drug interactions at a molecular level, thereby enhancing the understanding of both therapeutic effects and potential adverse reactions.
One prominent approach in molecular modeling is the use of drug target profile representations, which characterize drugs and their interactions based on the genes they target. This methodology integrates statistical metrics derived from human protein-protein interaction networks and signaling pathways to assess interaction intensity, efficacy, and action range between drug pairs. The application of an l2-regularized logistic regression model on these profiles has shown improved performance in predicting drug-drug interactions compared to traditional data integration methods, highlighting the importance of shared target genes and their connectivity in interaction networks (Mei & Zhang, 2021) [3].
Additionally, the development of geometric molecular graph representation learning models further advances the prediction of drug-drug interactions. These models focus on the covalent and non-covalent bond information of drug molecules, leveraging pre-training techniques to enhance the learning of molecular representations. This innovative approach has demonstrated superior predictive capabilities for new drug interactions, addressing the limitations of previous methods that primarily relied on established drug interaction networks (Jiang et al., 2024) [4].
Moreover, molecular docking and molecular dynamics simulations are critical tools in the optimization of pharmacological profiles. Molecular docking allows for the exploration of ligand conformations within binding sites of macromolecular targets, estimating binding affinities and interactions. These simulations provide insights into the stability of ligand-protein complexes over time, enabling medicinal chemists to identify potential improvements in drug candidates (Eid et al., 2013) [24].
The integration of machine learning with molecular modeling has also emerged as a powerful strategy to predict pharmacokinetic profiles and optimize drug interactions. For instance, machine learning frameworks that combine physiological data with compound-specific properties can simulate how drugs behave in the human body, thereby predicting drug-drug interactions more accurately. This is particularly useful in early drug development stages, where identifying potential interactions can mitigate adverse effects and enhance therapeutic efficacy (Jia et al., 2025) [26].
Overall, the applications of molecular modeling in predicting drug interactions encompass a variety of techniques, including statistical modeling, graph-based representations, molecular docking, and machine learning. These methodologies not only facilitate the identification of drug interactions but also optimize the pharmacological profiles of drug candidates, ultimately advancing the drug discovery process and improving patient safety.
3.3 Off-Target Interaction Prediction
Molecular modeling plays a pivotal role in predicting drug interactions, particularly in the context of off-target interaction prediction, which is crucial for understanding both therapeutic efficacy and potential adverse effects of drug candidates. Off-target interactions refer to the unintended binding of drugs to proteins other than their intended targets, which can lead to side effects or toxicity.
The methodologies for predicting off-target interactions have evolved significantly, leveraging various computational approaches. For instance, ligand-based models are employed in drug discovery to provide early indications of potential off-target interactions linked to adverse effects. These models can be integrated into a panel that allows for the comparison and search for compounds with similar interaction profiles. However, traditional methods often lack valid measures of confidence in their predictions, typically offering only point estimates. To address this limitation, a methodology using Conformal Prediction has been developed, which provides confidence p-values for each class of predictions, enhancing the reliability of the results (Lampa et al., 2018) [27].
Furthermore, the application of deep learning algorithms has shown great promise in predicting biomolecular interactions. These algorithms can analyze large datasets to identify complex patterns and relationships, thereby enhancing the accuracy of predictions regarding both on-target and off-target interactions. By utilizing features such as sequence data, structural information, and functional annotations, deep learning models can reduce the time and cost associated with screening compounds that exhibit high binding affinity to specific targets (Wang et al., 2025) [28].
The chemical-protein interactome (CPI) framework also plays a significant role in off-target identification. This framework creates an interaction strength matrix that assesses how drugs interact with multiple human proteins, thus exploring unexpected drug-protein interactions. By employing various computational strategies such as docking and chemical structure comparison, researchers can effectively predict off-target interactions, contributing to a deeper understanding of a drug's polypharmacology and advancing drug repositioning efforts (Yang et al., 2011) [29].
In addition to these methodologies, artificial intelligence (AI) and machine learning (ML) approaches have been increasingly integrated into the drug discovery process. For example, a computational repurposing framework developed for small molecules combines AI/ML-based and chemical similarity-based target prediction methods with cross-species transcriptomics information. This approach has identified numerous off-target interactions, confirming many predictions through experimental validation (Rao et al., 2023) [30].
Moreover, large-scale off-target identification methods have been proposed, such as REMAP, which utilizes a dual regularized one-class collaborative filtering algorithm to explore the chemical and protein space comprehensively. This method has demonstrated the capability to predict off-target interactions across different gene families efficiently and has been validated with extensive datasets, underscoring its potential for drug repurposing and safety assessment (Lim et al., 2016) [31].
Overall, molecular modeling in drug interaction prediction, particularly concerning off-target interactions, is facilitated by a variety of advanced computational techniques. These methodologies not only enhance the efficiency of drug discovery but also improve the understanding of drug safety profiles, ultimately aiding in the development of safer and more effective therapeutic agents.
4 Integration with Experimental Data
4.1 Data Validation and Cross-Verification
Molecular modeling plays a crucial role in predicting drug interactions by employing computational techniques that integrate various types of data, thereby enhancing the accuracy and reliability of predictions. The approach typically involves the use of molecular simulations, machine learning models, and data fusion methods, which can collectively provide insights into the interactions between drugs and their targets.
In the context of predicting drug interactions, molecular modeling utilizes various data sources, including genomic, proteomic, and chemogenomic information. For instance, Wang et al. (2011) introduced a kernel-based method that integrates multiple types of data to predict drug-protein interactions. This method involves characterizing drug-target interactions through the collection of pharmacological effects, chemical structures, and genomic information, which are then fused using a kernel function within a support vector machine (SVM)-based predictor. The study demonstrated that integrating diverse data sources significantly improves prediction accuracy compared to methods relying on single data sources[32].
Moreover, the integration of computational modeling with experimental data is essential for validating predictions and ensuring their applicability in drug discovery. Cichonska et al. (2017) proposed a systematic computational-experimental framework for predicting drug-target interactions. This framework utilized a kernel-based regression algorithm to predict binding affinities, which were subsequently validated through experimental testing of compound-kinase pairs. The results showed a high correlation (0.77, p < 0.0001) between predicted and measured bioactivities, underscoring the practical benefits of the model in filling gaps in existing interaction maps[33].
Additionally, the use of molecular simulations can extract structural and energetic information that complements experimental observations. Gago (2012) highlighted that advancements in molecular mechanics and computational technologies have enabled detailed studies of drug-receptor complexes. These simulations provide insights that may not be accessible through experimental methods alone, thereby aiding in the interpretation of biochemical results and guiding the design of new experiments[34].
Furthermore, Jiang et al. (2024) developed a geometric molecular graph representation learning model for predicting drug-drug interactions, which emphasizes the structural information of molecules. This model uses pre-training techniques to learn drug molecular representations, allowing for effective predictions of interactions, particularly for new drugs lacking prior binding profile information[4].
In summary, molecular modeling predicts drug interactions through the integration of diverse data sources and computational methods, validated by experimental data. This synergy enhances the understanding of drug-target and drug-drug interactions, ultimately contributing to more effective drug discovery and development processes. The combination of computational predictions with rigorous experimental validation is crucial for ensuring the reliability and applicability of the findings in clinical settings.
4.2 Case Studies of Successful Integration
Molecular modeling serves as a pivotal tool in predicting drug interactions, integrating computational techniques with experimental data to enhance the understanding of molecular behavior and interactions. The predictive capabilities of molecular modeling stem from its ability to simulate and analyze the interactions between drug molecules and their biological targets at a molecular level.
The integration of molecular modeling with experimental data has proven to be beneficial in various aspects of drug discovery and development. For instance, pharmacoinformatics combines quantum chemical methods, molecular modeling, and artificial intelligence (AI) to streamline the formulation development process by providing insights into molecular behavior and interaction mechanisms. This approach allows researchers to predict drug-excipient interactions, which are critical for ensuring the stability and biopharmaceutical performance of drug formulations [35].
In the realm of drug-drug interactions (DDIs), several studies highlight the efficacy of molecular modeling. For example, a machine learning framework was developed to predict DDIs by investigating the associations between genes targeted by two drugs. The study utilized a simple drug target profile representation to depict drugs and drug pairs, ultimately leading to the development of an l2-regularized logistic regression model. This model was validated through large-scale empirical studies, demonstrating that the framework outperformed existing data integration methods. The statistical metrics defined in this study indicated that interactions are more likely when drugs target common genes or when their target genes are connected through short paths in protein-protein interaction networks [3].
Moreover, a geometric molecular graph representation learning model (Mol-DDI) was proposed to predict DDIs based on molecular structures, focusing on covalent and non-covalent bond information. This model leverages the pre-training of large-scale models to enhance drug molecular representations and predict interactions during the fine-tuning process. Experimental results showed that the Mol-DDI model outperformed existing methods, particularly in predicting new drug interactions [4].
The successful integration of molecular modeling with experimental data is also evident in studies that explore drug-receptor interactions. Advances in molecular simulations and docking techniques allow for a detailed examination of drug-receptor complexes, enabling researchers to extract structural and energetic information that is often beyond the capabilities of experimental methods. This integration not only aids in the interpretation of biochemical results but also opens new avenues for research, guiding the design of new experiments [34].
In conclusion, molecular modeling predicts drug interactions by simulating the molecular behavior of drugs and their targets, integrating computational insights with experimental data to provide a comprehensive understanding of interaction mechanisms. The case studies presented demonstrate the effectiveness of this approach in enhancing the predictability of drug interactions, ultimately contributing to more efficient drug discovery and development processes.
5 Challenges and Limitations
5.1 Accuracy of Predictive Models
Molecular modeling plays a critical role in predicting drug interactions by simulating the interactions between drugs and their biological targets, typically proteins. The accuracy of these predictions is paramount, as it directly impacts drug efficacy and safety. However, several challenges and limitations affect the reliability of predictive models in this field.
The primary methods used in molecular modeling include molecular dynamics simulations, molecular docking, and free energy calculations. These approaches provide insights grounded in physical principles but often face significant hurdles. For instance, conventional physics-based computational methods can be computationally expensive, limited in scalability for larger systems, and may exhibit questionable predictive accuracy in real-world scenarios. These limitations necessitate the integration of advanced methodologies, such as deep learning (DL), which has emerged as a complementary tool to enhance predictive capabilities. DL techniques have been applied to augment molecular dynamics, improve molecular docking, and facilitate end-to-end modeling of protein-ligand interactions, among other applications [36].
Despite the advances in computational techniques, challenges remain in accurately predicting drug interactions. For instance, existing machine learning frameworks that predict drug-drug interactions often struggle with high model complexity and may not adequately elucidate the underlying molecular mechanisms. A study proposed a simplified drug target profile representation to predict interactions based on gene associations targeted by drugs, demonstrating that drugs targeting common genes or connected through short paths in protein-protein interaction networks are more likely to interact [3]. However, this approach still contends with the challenge of maintaining biological interpretability while ensuring model performance.
Moreover, predicting new drug interactions presents another layer of complexity. Traditional models often rely on established drug-drug interaction (DDI) networks, which can limit the discovery of novel interactions. A geometric molecular graph representation learning model was introduced to predict DDIs by focusing solely on molecular structure information, which showed improved performance in identifying new interactions [4].
The accuracy of predictive models is also influenced by various factors, including the choice of algorithms, the quality and size of the datasets used for training, and the biological relevance of the features incorporated into the models. For instance, models based on quantitative structure-activity relationships (QSAR) have been employed to predict drug-enzyme interactions, achieving high accuracy by leveraging extensive datasets [37]. However, the integration of genomic and network data can introduce challenges related to dimensionality and complexity, potentially impacting the robustness of predictions [38].
In summary, while molecular modeling offers significant potential for predicting drug interactions, it is not without its challenges. The integration of advanced computational techniques, such as deep learning, along with careful consideration of model design and data quality, is essential to enhance the accuracy and reliability of these predictive models in drug discovery and development.
5.2 Structural Data Quality
Molecular modeling serves as a pivotal tool in predicting drug interactions by simulating the behavior of drug-receptor complexes and elucidating the underlying mechanisms of molecular interactions. However, the effectiveness of these predictions is heavily influenced by the quality of structural data utilized in the modeling process.
Accurate modeling of drug interactions relies on detailed structural information about both the drugs and their biological targets. The precision of molecular dynamics simulations, molecular docking, and other computational methods hinges on the availability of high-quality structural data, which is often derived from experimental techniques such as X-ray crystallography or nuclear magnetic resonance (NMR) spectroscopy. These methods provide crucial insights into the three-dimensional arrangements of atoms within biomolecules, which are essential for understanding how drugs bind to their targets.
Despite advancements in structural biology, challenges persist in obtaining high-resolution structural data. For instance, many proteins involved in drug interactions may exist in multiple conformations or may be part of larger complexes, complicating the interpretation of structural data. Furthermore, the dynamic nature of biomolecular interactions means that static structural snapshots may not fully capture the range of conformational states relevant to drug binding. This limitation can lead to inaccuracies in predicting binding affinities and interaction dynamics.
Additionally, the complexity of molecular interactions poses further challenges. Drug-drug interactions (DDIs) are influenced not only by the structural properties of the drugs but also by their interactions with various biological entities, including proteins, genes, and metabolic pathways. Conventional models often overlook these multifaceted relationships, focusing solely on structural features. Recent approaches, such as the integration of knowledge graphs with molecular graph representations, aim to incorporate both structural and semantic information, enhancing the predictive power of models [39].
The limitations of existing computational methods are evident in the context of drug-drug interaction prediction. Many current models rely on predefined drug interaction networks or structural features, which can restrict their ability to predict interactions for novel compounds. For instance, while deep learning-based models have shown promise in enhancing predictive accuracy, they still face challenges related to model complexity and the interpretability of results [3].
Moreover, the quality of structural data directly impacts the reliability of predictive models. Inconsistent or low-quality structural data can lead to erroneous predictions, emphasizing the need for robust validation methods and the integration of diverse data sources. Studies have demonstrated that incorporating biological insights, such as gene-drug associations and network-based features, can improve the understanding of drug interactions and mitigate some of the limitations associated with structural data [40].
In summary, while molecular modeling provides valuable insights into drug interactions, its predictive accuracy is contingent upon the quality of structural data. Ongoing advancements in structural biology, coupled with innovative computational approaches, are essential to overcoming these challenges and enhancing the reliability of drug interaction predictions.
5.3 Computational Resource Constraints
Molecular modeling serves as a pivotal tool in predicting drug interactions, particularly in the context of rational drug discovery. This approach encompasses a variety of computational techniques that facilitate the understanding of how drugs interact with their biological targets at the molecular level. Key methodologies include molecular dynamics simulations, molecular docking, and free energy calculations, which collectively provide insights into the binding affinities and mechanisms of action of drug candidates.
Despite the advantages offered by molecular modeling, significant challenges and limitations persist, particularly regarding computational resource constraints. Traditional physics-based computational methods, while theoretically rigorous, often suffer from high computational costs and limited scalability. For instance, the deployment of molecular dynamics simulations can be hampered by the extensive computational power required to simulate large biological systems accurately. These limitations can restrict the practical application of such methods in real-world drug discovery scenarios, where the ability to rapidly screen and evaluate numerous compounds is essential (Wang et al., 2025) [36].
Moreover, the integration of deep learning techniques into molecular modeling has shown promise in addressing some of these challenges. Deep learning can augment traditional methods by providing data-driven insights that enhance predictive accuracy and efficiency. However, the transition to these advanced methodologies also necessitates substantial computational resources and expertise, which may not be readily available in all research settings (Hasan et al., 2022) [25].
In the context of drug interaction prediction, the need for high-throughput capabilities is crucial. The computational expense associated with molecular dynamics and other physics-based methods can limit their application in extensive drug screening processes. Consequently, researchers are increasingly exploring hybrid approaches that combine computational modeling with experimental data to optimize the drug discovery pipeline (Narykov et al., 2023) [41].
In summary, while molecular modeling is a powerful tool for predicting drug interactions, its effectiveness is significantly influenced by computational resource constraints. The ongoing development of more efficient algorithms and the integration of machine learning techniques may offer pathways to mitigate these challenges, thereby enhancing the capabilities of molecular modeling in drug discovery.
6 Future Perspectives
6.1 Advances in Computational Techniques
Molecular modeling serves as a pivotal tool in predicting drug interactions, leveraging computational techniques to enhance our understanding of how drugs interact with biological systems. The integration of advanced methodologies such as molecular dynamics simulations, quantitative structure-activity relationship (QSAR) models, and artificial intelligence (AI) has significantly improved the predictive capabilities of molecular modeling in pharmacology.
Recent advances in molecular simulations allow researchers to explore drug interactions at a molecular level, offering insights into the binding affinities and conformational dynamics of drug-receptor complexes. For instance, the evolution of molecular mechanics force fields and improvements in computational technologies, including processing speeds and data-storage capacity, have facilitated detailed simulations that provide structural and energetic information about drug interactions that are often beyond the reach of experimental methods (Gago 2012) [34].
In the context of drug transport across biological membranes, molecular dynamics simulations have proven invaluable. These simulations enable the prediction of drug solubility and the characterization of drug transport pathways, effectively allowing researchers to assess how hydrophobic and hydrophilic solutes interact with membranes (Loverde 2014) [11]. This capability is crucial for understanding pharmacokinetics and the overall efficacy of drug formulations.
The incorporation of AI in molecular modeling has further revolutionized the prediction of drug interactions. AI-driven models can analyze vast datasets to identify patterns and relationships between drug structures and their biological effects. For example, machine learning techniques have been employed to model drug metabolism, allowing for predictions regarding the metabolic fate of compounds based on their chemical structures (Fox & Kriegl 2006) [42]. These predictive models enhance the drug discovery process by identifying potential drug candidates and assessing their interactions with metabolic enzymes.
Moreover, computational systems biology approaches integrate omics data from various biological layers to predict molecular interactions and drug responses, thus addressing the complexity of disease mechanisms (Yue & Dutta 2022) [43]. By leveraging these models, researchers can optimize drug combinations and tailor therapies to individual patient profiles, ultimately improving therapeutic outcomes.
Looking forward, the ongoing refinement of computational modeling techniques is expected to enhance the prediction of drug interactions significantly. The development of multi-target machine learning models, such as those used for predicting drug-enzyme interactions, represents a promising direction in this field (Concu et al. 2023) [37]. As computational methods continue to evolve, they will likely become even more integral to drug discovery and development, enabling researchers to navigate the complexities of pharmacology with greater precision and efficiency.
In summary, molecular modeling predicts drug interactions through a combination of advanced simulations, machine learning techniques, and systems biology approaches, paving the way for more effective drug discovery and personalized medicine strategies. The future of this field appears bright, with continuous advancements poised to address current challenges and improve the safety and efficacy of therapeutic interventions.
6.2 Role of Artificial Intelligence in Molecular Modeling
Molecular modeling serves as a pivotal tool in predicting drug interactions by utilizing computational techniques to simulate and analyze the interactions between drugs and their biological targets. The advent of artificial intelligence (AI) has significantly enhanced the capabilities of molecular modeling, allowing for more accurate and efficient predictions in pharmacology.
In the context of cardiovascular pharmacology, recent advancements in molecular simulations and big data analysis have opened new avenues for rationalizing drug interactions with pharmacological targets. AI has emerged as a powerful tool that can analyze large-scale biological data, identify molecular targets, and elucidate pathways that advance pharmacological knowledge. Despite the promising potential, the application of these computational approaches in cardiovascular diseases and drug discovery has not progressed uniformly across different pharmacological fields, indicating a need for further integration of AI in molecular modeling[1].
AI algorithms are particularly effective in analyzing complex datasets that encompass various aspects of drug interactions, including chemical structures, pharmacological properties, and known interaction patterns. By integrating this multifaceted information, AI can provide mechanistic insights and identify potential risks associated with drug interactions. This capability is crucial, especially when assessing interactions between conventional drugs and natural products such as herbal supplements, which often have complex mixtures and limited pharmacological data[44].
The incorporation of AI into drug-target interaction prediction and lead optimization is revolutionizing pharmaceutical research. AI techniques enhance the prediction accuracy of drug interactions by systematically modeling relationships among drugs, targets, and diseases. This not only accelerates the discovery timelines but also reduces costs associated with traditional trial-and-error methods. Recent reviews have highlighted the potential of deep learning tools in guiding researchers through various phases of drug discovery, including target identification and synthetic accessibility prediction[45].
Moreover, AI-driven methods have shown promise in predicting biomolecular interactions, which is essential for understanding drug mechanisms and side effects. By leveraging deep learning algorithms, researchers can analyze large datasets, reducing the time and cost of identifying compounds with high binding affinity to target molecules. These models also facilitate the identification of key residues involved in drug interactions, thereby improving the understanding of drug mechanisms and enhancing the overall drug development process[28].
Looking ahead, the integration of AI into molecular modeling is poised to address several challenges in drug discovery. As AI continues to evolve, it is expected to enhance the predictability of drug interactions, facilitate the design of multi-target drugs, and ultimately contribute to personalized medicine strategies. The ongoing collaboration between computational and domain experts will be critical in realizing the full potential of AI in molecular modeling and drug interaction prediction, thereby advancing the field of pharmacology[46].
In summary, molecular modeling, bolstered by AI technologies, is transforming the landscape of drug interaction prediction. By harnessing the power of AI, researchers can achieve more accurate, efficient, and insightful predictions, paving the way for innovative drug discovery and development approaches.
7 Conclusion
The exploration of molecular modeling techniques reveals significant advancements in predicting drug interactions, particularly through methods such as molecular docking, molecular dynamics simulations, and quantitative structure-activity relationship (QSAR) modeling. These methodologies have enabled researchers to rationalize drug design, optimize pharmacological profiles, and predict off-target interactions, ultimately enhancing the efficiency of drug discovery processes. However, challenges persist, including the accuracy of predictive models, the quality of structural data, and computational resource constraints. Future research directions should focus on integrating artificial intelligence to improve predictive capabilities, refining existing models, and enhancing the reliability of predictions through experimental validation. The ongoing evolution of computational techniques promises to address these challenges, paving the way for more effective and safer therapeutic agents in the pharmaceutical landscape.
References
- [1] Jennifer Lagoutte-Renosi;Florentin Allemand;Christophe Ramseyer;Semen Yesylevskyy;Siamak Davani. Molecular modeling in cardiovascular pharmacology: Current state of the art and perspectives.. Drug discovery today(IF=7.5). 2022. PMID:34863931. DOI: 10.1016/j.drudis.2021.11.026.
- [2] Mohammed Maniruzzaman;Jiayun Pang;David J Morgan;Dennis Douroumis. Molecular modeling as a predictive tool for the development of solid dispersions.. Molecular pharmaceutics(IF=4.5). 2015. PMID:25734898. DOI: 10.1021/mp500510m.
- [3] Suyu Mei;Kun Zhang. A machine learning framework for predicting drug-drug interactions.. Scientific reports(IF=3.9). 2021. PMID:34475500. DOI: 10.1038/s41598-021-97193-8.
- [4] Zhenyu Jiang;Pingjian Ding;Cong Shen;Xiaopeng Dai. Geometric Molecular Graph Representation Learning Model for Drug-Drug Interactions Prediction.. IEEE journal of biomedical and health informatics(IF=6.8). 2024. PMID:39226203. DOI: 10.1109/JBHI.2024.3453956.
- [5] Ahmad Abu Turab Naqvi;Taj Mohammad;Gulam Mustafa Hasan;Md Imtaiyaz Hassan. Advancements in Docking and Molecular Dynamics Simulations Towards Ligand-receptor Interactions and Structure-function Relationships.. Current topics in medicinal chemistry(IF=3.3). 2018. PMID:30360721. DOI: 10.2174/1568026618666181025114157.
- [6] Nataraj S Pagadala;Khajamohiddin Syed;Jack Tuszynski. Software for molecular docking: a review.. Biophysical reviews(IF=3.7). 2017. PMID:28510083. DOI: 10.1007/s12551-016-0247-1.
- [7] John Lee;Canh Hao Nguyen;Hiroshi Mamitsuka. Beyond rigid docking: deep learning approaches for fully flexible protein-ligand interactions.. Briefings in bioinformatics(IF=7.7). 2025. PMID:40900115. DOI: 10.1093/bib/bbaf454.
- [8] P C Agu;C A Afiukwa;O U Orji;E M Ezeh;I H Ofoke;C O Ogbu;E I Ugwuja;P M Aja. Molecular docking as a tool for the discovery of molecular targets of nutraceuticals in diseases management.. Scientific reports(IF=3.9). 2023. PMID:37592012. DOI: 10.1038/s41598-023-40160-2.
- [9] Edward King;Erick Aitchison;Han Li;Ray Luo. Recent Developments in Free Energy Calculations for Drug Discovery.. Frontiers in molecular biosciences(IF=4.0). 2021. PMID:34458321. DOI: 10.3389/fmolb.2021.712085.
- [10] Begüm Sarac;Seydanur Yücer;Fatih Ciftci;Mansour Ghorbanpour;Esma Ahlatcioglu Ozerol. Molecular Dynamics Simulations and Their Novel Applications in Drug Delivery for Cancer Treatment: A Review.. Annals of biomedical engineering(IF=5.4). 2025. PMID:41023346. DOI: 10.1007/s10439-025-03864-2.
- [11] Sharon M Loverde. Molecular Simulation of the Transport of Drugs across Model Membranes.. The journal of physical chemistry letters(IF=4.6). 2014. PMID:26270362. DOI: 10.1021/jz500321d.
- [12] Apurba Bhattarai;Yinglong Miao. Gaussian accelerated molecular dynamics for elucidation of drug pathways.. Expert opinion on drug discovery(IF=4.9). 2018. PMID:30371112. DOI: 10.1080/17460441.2018.1538207.
- [13] Xuewei Liu;Danfeng Shi;Shuangyan Zhou;Hongli Liu;Huanxiang Liu;Xiaojun Yao. Molecular dynamics simulations and novel drug discovery.. Expert opinion on drug discovery(IF=4.9). 2018. PMID:29139324. DOI: 10.1080/17460441.2018.1403419.
- [14] David W Borhani;David E Shaw. The future of molecular dynamics simulations in drug discovery.. Journal of computer-aided molecular design(IF=3.1). 2012. PMID:22183577. DOI: 10.1007/s10822-011-9517-y.
- [15] Attilio V Vargiu;Alessandra Magistrato. Atomistic-level portrayal of drug-DNA Interplay: a history of courtships and meetings revealed by molecular simulations.. ChemMedChem(IF=3.4). 2014. PMID:25099999. DOI: 10.1002/cmdc.201402203.
- [16] Markus A Lill. Multi-dimensional QSAR in drug discovery.. Drug discovery today(IF=7.5). 2007. PMID:18061879. DOI: 10.1016/j.drudis.2007.08.004.
- [17] Sarfaraz K Niazi;Zamara Mariam. Recent Advances in Machine-Learning-Based Chemoinformatics: A Comprehensive Review.. International journal of molecular sciences(IF=4.9). 2023. PMID:37511247. DOI: 10.3390/ijms241411488.
- [18] Gyoung Jin Park;Nam Sook Kang. ADis-QSAR: a machine learning model based on biological activity differences of compounds.. Journal of computer-aided molecular design(IF=3.1). 2023. PMID:37382799. DOI: 10.1007/s10822-023-00517-1.
- [19] Ivan Rusyn;Alexander Sedykh;Yen Low;Kathryn Z Guyton;Alexander Tropsha. Predictive modeling of chemical hazard by integrating numerical descriptors of chemical structures and short-term toxicity assay data.. Toxicological sciences : an official journal of the Society of Toxicology(IF=4.1). 2012. PMID:22387746. DOI: 10.1093/toxsci/kfs095.
- [20] Joyce K Daré;Teodorico C Ramalho;Matheus P Freitas. 3D perspective into MIA-QSAR: A case for anti-HCV agents.. Chemical biology & drug design(IF=3.3). 2019. PMID:30411494. DOI: 10.1111/cbdd.13440.
- [21] Alejandro Speck-Planche;M N D S Cordeiro. Multi-Target QSAR Approaches for Modeling Protein Inhibitors. Simultaneous Prediction of Activities Against Biomacromolecules Present in Gram-Negative Bacteria.. Current topics in medicinal chemistry(IF=3.3). 2015. PMID:25961517. DOI: 10.2174/1568026615666150506144814.
- [22] Age Aleksander Skjevik;Knut Teigen;Aurora Martinez. Overview of computational methods employed in early-stage drug discovery.. Future medicinal chemistry(IF=3.4). 2009. PMID:21426070. DOI: 10.4155/fmc.09.7.
- [23] Zarko Gagic;Dusan Ruzic;Nemanja Djokovic;Teodora Djikic;Katarina Nikolic. In silico Methods for Design of Kinase Inhibitors as Anticancer Drugs.. Frontiers in chemistry(IF=4.2). 2019. PMID:31970149. DOI: 10.3389/fchem.2019.00873.
- [24] Sameh Eid;Adam Zalewski;Martin Smieško;Beat Ernst;Angelo Vedani. A molecular-modeling toolbox aimed at bridging the gap between medicinal chemistry and computational sciences.. International journal of molecular sciences(IF=4.9). 2013. PMID:23344039. DOI: 10.3390/ijms14010684.
- [25] Md Rifat Hasan;Ahad Amer Alsaiari;Burhan Zain Fakhurji;Mohammad Habibur Rahman Molla;Amer H Asseri;Md Afsar Ahmed Sumon;Moon Nyeo Park;Foysal Ahammad;Bonglee Kim. Application of Mathematical Modeling and Computational Tools in the Modern Drug Design and Development Process.. Molecules (Basel, Switzerland)(IF=4.6). 2022. PMID:35807415. DOI: 10.3390/molecules27134169.
- [26] Xuelian Jia;Donato Teutonico;Saroj Dhakal;Yorgos M Psarellis;Alexandra Abos;Hao Zhu;Panteleimon D Mavroudis;Nikhil Pillai. Application of Machine Learning and Mechanistic Modeling to Predict Intravenous Pharmacokinetic Profiles in Humans.. Journal of medicinal chemistry(IF=6.8). 2025. PMID:40146185. DOI: 10.1021/acs.jmedchem.5c00340.
- [27] Samuel Lampa;Jonathan Alvarsson;Staffan Arvidsson Mc Shane;Arvid Berg;Ernst Ahlberg;Ola Spjuth. Predicting Off-Target Binding Profiles With Confidence Using Conformal Prediction.. Frontiers in pharmacology(IF=4.8). 2018. PMID:30459617. DOI: 10.3389/fphar.2018.01256.
- [28] Haoping Wang;Xiangjie Meng;Yang Zhang. Biomolecular Interaction Prediction: The Era of AI.. Advanced science (Weinheim, Baden-Wurttemberg, Germany)(IF=14.1). 2025. PMID:40671265. DOI: 10.1002/advs.202509501.
- [29] Lun Yang;Ke-Jian Wang;Li-Shan Wang;Anil G Jegga;Sheng-Ying Qin;Guang He;Jian Chen;Yue Xiao;Lin He. Chemical-protein interactome and its application in off-target identification.. Interdisciplinary sciences, computational life sciences(IF=3.9). 2011. PMID:21369884. DOI: 10.1007/s12539-011-0051-8.
- [30] Mohan Rao;Eric McDuffie;Clifford Sachs. Artificial Intelligence/Machine Learning-Driven Small Molecule Repurposing via Off-Target Prediction and Transcriptomics.. Toxics(IF=4.1). 2023. PMID:37888725. DOI: 10.3390/toxics11100875.
- [31] Hansaim Lim;Aleksandar Poleksic;Yuan Yao;Hanghang Tong;Di He;Luke Zhuang;Patrick Meng;Lei Xie. Large-Scale Off-Target Identification Using Fast and Accurate Dual Regularized One-Class Collaborative Filtering and Its Application to Drug Repurposing.. PLoS computational biology(IF=3.6). 2016. PMID:27716836. DOI: 10.1371/journal.pcbi.1005135.
- [32] Yong-Cui Wang;Chun-Hua Zhang;Nai-Yang Deng;Yong Wang. Kernel-based data fusion improves the drug-protein interaction prediction.. Computational biology and chemistry(IF=3.1). 2011. PMID:22099632. DOI: 10.1016/j.compbiolchem.2011.10.003.
- [33] Anna Cichonska;Balaguru Ravikumar;Elina Parri;Sanna Timonen;Tapio Pahikkala;Antti Airola;Krister Wennerberg;Juho Rousu;Tero Aittokallio. Computational-experimental approach to drug-target interaction mapping: A case study on kinase inhibitors.. PLoS computational biology(IF=3.6). 2017. PMID:28787438. DOI: 10.1371/journal.pcbi.1005678.
- [34] Federico Gago. Molecular simulations of drug-receptor complexes in anticancer research.. Future medicinal chemistry(IF=3.4). 2012. PMID:23088276. DOI: 10.4155/fmc.12.149.
- [35] Sourav Chougule;Kanika Manchanda;Mehak Sood;Arvind K Bansal;Prasad V Bharatam. Pharmacoinformatics in Drug-Excipient Interactions.. Molecular pharmaceutics(IF=4.5). 2025. PMID:41144649. DOI: 10.1021/acs.molpharmaceut.5c00801.
- [36] Yuzhe Wang;Yibo Li;Jiaxiao Chen;Luhua Lai. Modeling protein-ligand interactions for drug discovery in the era of deep learning.. Chemical Society reviews(IF=39.0). 2025. PMID:41117015. DOI: 10.1039/d5cs00415b.
- [37] Riccardo Concu;Maria Natália Dias Soeiro Cordeiro;Martín Pérez-Pérez;Florentino Fdez-Riverola. MOZART, a QSAR Multi-Target Web-Based Tool to Predict Multiple Drug-Enzyme Interactions.. Molecules (Basel, Switzerland)(IF=4.6). 2023. PMID:36770857. DOI: 10.3390/molecules28031182.
- [38] Zachary Stanfield;Mustafa Coşkun;Mehmet Koyutürk. Drug Response Prediction as a Link Prediction Problem.. Scientific reports(IF=3.9). 2017. PMID:28067293. DOI: 10.1038/srep40321.
- [39] Yu Li;Zhu-Hong You;Yuan Yang;Cheng-Gang Mi. KGMAEDDI: Knowledge Graph and Molecular-Graph Masked Autoencoder for Drug-Drug Interaction Prediction.. IEEE journal of biomedical and health informatics(IF=6.8). 2025. PMID:40966139. DOI: 10.1109/JBHI.2025.3610870.
- [40] Siqi Chen;Tiancheng Li;Luna Yang;Fei Zhai;Xiwei Jiang;Rongwu Xiang;Guixia Ling. Artificial intelligence-driven prediction of multiple drug interactions.. Briefings in bioinformatics(IF=7.7). 2022. PMID:36168896. DOI: 10.1093/bib/bbac427.
- [41] Oleksandr Narykov;Yitan Zhu;Thomas Brettin;Yvonne A Evrard;Alexander Partin;Maulik Shukla;Fangfang Xia;Austin Clyde;Priyanka Vasanthakumari;James H Doroshow;Rick L Stevens. Integration of Computational Docking into Anti-Cancer Drug Response Prediction Models.. Cancers(IF=4.4). 2023. PMID:38201477. DOI: 10.3390/cancers16010050.
- [42] Thomas Fox;Jan M Kriegl. Machine learning techniques for in silico modeling of drug metabolism.. Current topics in medicinal chemistry(IF=3.3). 2006. PMID:16918470. DOI: 10.2174/156802606778108915.
- [43] Rongting Yue;Abhishek Dutta. Computational systems biology in disease modeling and control, review and perspectives.. NPJ systems biology and applications(IF=3.5). 2022. PMID:36192551. DOI: 10.1038/s41540-022-00247-4.
- [44] Marios Spanakis;Eleftheria Tzamali;Georgios Tzedakis;Chryssalenia Koumpouzi;Matthew Pediaditis;Aristides Tsatsakis;Vangelis Sakkalis. Artificial Intelligence Models and Tools for the Assessment of Drug-Herb Interactions.. Pharmaceuticals (Basel, Switzerland)(IF=4.8). 2025. PMID:40143062. DOI: 10.3390/ph18030282.
- [45] Qiqi Wang;Boyan Sun;Yunpeng Yi;Tony Velkov;Jianzhong Shen;Chongshan Dai;Haiyang Jiang. Progress of AI-Driven Drug-Target Interaction Prediction and Lead Optimization.. International journal of molecular sciences(IF=4.9). 2025. PMID:41155330. DOI: 10.3390/ijms262010037.
- [46] Anna Cichońska;Balaguru Ravikumar;Rayees Rahman. AI for targeted polypharmacology: The next frontier in drug discovery.. Current opinion in structural biology(IF=7.0). 2024. PMID:38215530. DOI: 10.1016/j.sbi.2023.102771.
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
Molecular Modeling · Drug Interactions · Computational Techniques · Drug Design · Artificial Intelligence
© 2025 MaltSci
