View Article

Abstract

Artificial intelligence (AI) is emerging as one of the most influential technologies in healthcare, with pharmacology standing at the forefront of its applications. By harnessing large and complex datasets, AI has accelerated drug discovery, improved clinical trial efficiency, enabled model-informed precision dosing, and provided new insights into pharmacogenomics and adverse drug reaction prediction. Recent advances, such as deep learning models for protein structure prediction and reinforcement learning for individualized dosing strategies, illustrate the potential of AI to transform every stage of the drug development and utilization cycle.At the same time, challenges remain significant. Issues of algorithmic transparency, dataset quality, bias, and hidden stratification continue to affect reproducibility and clinical trust. Regulatory frameworks are still evolving, and ethical concerns surrounding accountability, privacy, and equity demand urgent attention. The integration of AI into pharmacology is further complicated by infrastructural limitations in low- and middle-income countries, which risk widening existing disparities in healthcare delivery. Nevertheless, opportunities for innovation are vast. Blockchain technologies promise secure and transparent data exchange; federated learning enables collaborative modeling without compromising patient privacy; and integration with precision medicine holds the promise of tailored therapies based on genetic and phenotypic profiles. These developments point toward a future where AI is not a mere tool but a foundational element of pharmacological research and practice. Overall, AI has demonstrated the capacity to improve efficiency, accuracy, and personalization in pharmacology, but its long-term success will depend on inclusive datasets, robust governance, and interdisciplinary collaboration. With careful implementation, AI can move from promise to practice, shaping a safer and more innovative future.

Keywords

Artificial intelligence, Machine learning, Drug discovery, Pharmacovigilance, Precision medicine

Introduction

Artificial intelligence (AI) has rapidly evolved from a theoretical concept in computer science into a transformative tool across healthcare, including pharmacology. The capacity of AI to process complex, multidimensional datasets, recognize hidden patterns, and generate predictive insights makes it uniquely suited for pharmacological applications where the traditional “trial-and-error” approach is time-consuming, costly, and often limited by human bias. The convergence of computational power, availability of biomedical big data, and advances in machine learning algorithms has opened new possibilities for drug discovery, personalized therapy, clinical trial optimization, pharmacovigilance, and regulatory science (1,2) Pharmacology has historically relied on experimental and observational science to determine drug mechanisms, safety, and efficacy. With the exponential growth of data from genomics, proteomics, metabolomics, imaging, electronic health records (EHRs), and real-world evidence, the need for robust analytical tools has become urgent. AI provides solutions by integrating diverse datasets to accelerate therapeutic innovations while maintaining precision. As Ferguson et al. (2001) emphasized in their early review on technology integration in pharmacology, innovation must be grounded in rigorous evaluation, ethical considerations, and alignment with clinical needs.

This principle continues to guide the incorporation of AI into modern pharmacological research and practice. The integration of AI is not simply a matter of automating existing processes. Instead, it represents a paradigm shift—transforming how pharmacologists conceptualize targets, design molecules, predict pharmacokinetics, monitor adverse effects, and tailor treatments to individual patients. Despite the enthusiasm, challenges persist, including issues of transparency, validation, reproducibility, bias, and regulation (3,4) . Thus, a critical review of AI in pharmacology must balance optimism with scientific caution, addressing both current achievements and unresolved barriers. This article aims to provide a comprehensive account of AI in pharmacology: tracing its conceptual foundations, describing how AI is integrated across drug discovery, clinical pharmacology, and pharmacovigilance, critically appraising current achievements, identifying systemic limitations, and projecting future directions such as blockchain-enabled AI, digital twins, and federated learning.

1. FUNDAMENTALS OF ARTIFICIAL INTELLIGENCE

1.1 Defining Artificial Intelligence

Artificial intelligence refers to the simulation of human cognitive functions such as learning, reasoning, problem-solving, and decision-making by machines. In the healthcare context, AI systems are designed to analyze data, recognize patterns, and generate actionable insights. Unlike traditional statistical approaches, AI can handle nonlinear, high-dimensional datasets without explicit preprogrammed rules (5).

AI encompasses several branches:

  • Machine Learning (ML): Algorithms that learn from data and improve over time.
  • Deep Learning (DL): Subset of ML using neural networks with multiple layers, capable of complex feature extraction.
  • Reinforcement Learning (RL): Algorithms that optimize decision-making based on trial-and-error feedback.
  • Natural Language Processing (NLP): Techniques that analyze unstructured biomedical texts and clinical narratives (6,7)

1.2 Historical Development of AI in Medicine

The early AI systems in healthcare were expert systems (e.g., MYCIN in the 1970s), which relied on rule-based logic to guide clinical decisions. However, these systems were limited by their rigidity and inability to scale with increasing data. The 21st century witnessed a paradigm shift with the advent of machine learning and deep learning, supported by improved computational resources and access to large biomedical datasets (8)

In pharmacology, early applications included computer-assisted drug design and predictive toxicology. Today, AI spans the entire pharmacological spectrum, from molecular docking to predicting real-world drug effectiveness and safety.

1.3 Core AI Methodologies Relevant to Pharmacology

Supervised Learning: Models trained on labeled datasets to predict outcomes (e.g., drug response prediction using genetic markers).

Unsupervised Learning: Identifies hidden clusters and associations in unlabeled datasets (e.g., novel adverse event detection).

Deep Neural Networks: Including convolutional neural networks (CNNs) for image-based drug screening and recurrent neural networks (RNNs) for sequential data such as longitudinal EHRs. The introduction of transformer models (9) and their derivatives have enabled breakthroughs in sequence analysis, protein folding, and biomedical NLP.

Generative Models: Generative adversarial networks (GANs) and variational autoencoders (VAEs) are applied to de novo drug design, virtual screening, and missing data imputation (10).

Reinforcement Learning: Increasingly important in precision dosing and adaptive clinical trial design (11).

1.4 Data Sources for AI in Pharmacology

AI in pharmacology thrives on data availability. Key sources include:

Omics data (genomics, proteomics, metabolomics, transcriptomics): Driving pharmacogenomics and systems pharmacology.

Biomedical imaging (radiomics and pathomics): Supporting drug response prediction and biomarker discovery (12).

Electronic Health Records (EHRs): Providing longitudinal data on drug efficacy, safety, and patient adherence (13).

Pharmacovigilance Databases: AI-assisted monitoring of spontaneous reports and social media mining for adverse drug reactions (14,15) .

Clinical Trials and Real-World Evidence: AI applications in protocol optimization and regulatory submissions (16,17).

1.5 Distinguishing AI from Conventional Computational Pharmacology

Conventional computational pharmacology tools, such as molecular docking and quantitative structure–activity relationship (QSAR) models, are deterministic and hypothesis-driven . AI, in contrast, is hypothesis-generating, able to uncover unexpected associations and emergent patterns without explicit rules. For example, whereas QSAR models require pre-specified descriptors, deep learning approaches can autonomously learn relevant features from raw chemical structures (18) .

1.6 Ethical Foundations in AI Development

Before discussing integration, it is crucial to highlight the ethical framework guiding AI deployment. As the World Medical Association (19) emphasized, AI should be viewed as “augmented intelligence,” assisting clinicians and pharmacologists rather than replacing them. Transparency, fairness, and accountability are foundational to responsible AI.

The conceptual development of artificial intelligence in medicine has its origins in early statistical learning and clinical decision-support systems. Early applications of regression-based models and pattern recognition in (20) biomedical research demonstrated the potential of computers to mimic aspects of human reasoning in healthcare contexts. These pioneering steps laid the foundation for modern AI, particularly when integrated with large-scale electronic health data.

The introduction of deep learning in healthcare marked a major inflection point. For example, neural architectures trained on longitudinal patient records demonstrated that EHR data could predict hospitalization risk, mortality, and drug side effects with remarkable accuracy (8,21) . These studies underscored the transition from handcrafted features to end-to-end learning, an approach that is now central to pharmacological applications.

In parallel, computational chemistry and pharmacology benefitted from machine learning methods even before “AI” became a popular term.(22) Techniques such as quantitative structure–activity relationship (QSAR) modeling were among the first to highlight the ability of algorithms to extract hidden patterns linking molecular descriptors with pharmacological outcomes (23). Advances in QSAR later merged with deep generative models, producing drug design strategies that go beyond traditional high-throughput screening.

The emphasis on interpretability and biological plausibility remains critical. As Handelman et al. (2018) argue, AI models must not only achieve predictive accuracy but also generate mechanistic insights to be truly transformative in pharmacology. For this reason, early experiences with predictive toxicology and metabolic modeling continue to inform how pharmacologists evaluate the validity of newer neural-network–based models.

Figure 1: AI Applications Across the Drug Discovery and Development Pipeline. (24)

Table 1 (concise). AI methods and where they’re used in pharmacology

Method

Typical data

Key uses in pharmacology

Supervised ML

EHR, labs, trials

Response/risk prediction; ADR detection aids

Deep learning (CNN/RNN/Transformers)

Images, sequences, longitudinal EHR

Radiomics/pathomics; sequence modeling; representation learning

Generative models (VAEs/GANs/diffusion; GAIN)

Chemical graphs/ SMILES; omics; tabular

De novo design; virtual screening; missing-data imputation

Knowledge graphs / networks

Literature + DrugBank/ ChEMBL/ pathways

Target ID; drug repurposing; DDI/ADR links

RL & model-informed precision dosing (MIPD)

PK/PD streams; TDM

Adaptive/individualized dosing; regimen optimization

Hybrid PK–ML

Sparse concentrations + covariates

Exposure prediction (e.g., vanco, tacrolimus, isavuconazole)

NLP (clinical & social)

Notes, discharge summaries, social media

ADR extraction; DDI mining; signal triage

Trial & regulatory analytics

Historic trials, registries

Eligibility screening; early stop/site risk models; reporting standards

Federated / privacy-preserving ML

Multi-site EHR/omics

Cross-institution modeling without data pooling

2. INTEGRATION OF ARTIFICIAL INTELLIGENCE IN PHARMACOLOGY

2.1 AI in Drug Discovery and Development

Drug discovery traditionally involves labor-intensive experimental screening of thousands of compounds, with high costs and low success rates. Artificial intelligence offers a paradigm shift by enabling rational drug design, target identification, virtual screening, and toxicity prediction.

One of the most transformative applications is de novo drug design, where generative models propose novel molecular structures with optimized pharmacological properties. Deep learning–based platforms such as DeepAffinity and DeepTox have demonstrated high accuracy in predicting compound–protein binding affinities and toxicity, respectively0 (18,25).  AI-powered structure prediction, epitomized by AlphaFold (26) has revolutionized protein modeling, enabling pharmacologists to design drugs against previously “undruggable” targets.

Drug repurposing represents another AI success story. During the COVID-19 pandemic, AI-driven approaches accelerated candidate selection, including baricitinib as a treatment option (27). Knowledge graph–based approaches and systems pharmacology pipelines systematically integrate heterogeneous biomedical data to prioritize repurposing candidates (28).

Moreover, AI enhances virtual high-throughput screening by learning from chemical libraries to predict hit-to-lead optimization. In oncology, for instance, Bayesian and neural network–based predictive models are used to forecast drug–target interactions and resistance patterns (29). These applications underline how AI compresses the timeline and cost of drug development while improving success rates (30,31)

2.2 AI in Clinical Pharmacology

2.2.1 Precision Dosing and PK/PD Modeling

Therapeutic drug monitoring and individualized dosing are longstanding goals of clinical pharmacology. AI, particularly reinforcement learning and hybrid pharmacokinetic (PK)–machine learning models, has enabled model-informed precision dosing (MIPD). Studies on vancomycin, isavuconazole, tacrolimus, and mycophenolic acid demonstrate that hybrid ML–PK models outperform conventional Bayesian approaches in predicting drug exposure (32–34).

For example, reinforcement learning frameworks have been applied to precision dosing of chemotherapeutics, where the algorithm adjusts dose regimens dynamically based on patient response data (11). Such models reduce toxicity risk while maximizing therapeutic benefit, especially in populations with high variability such as pediatrics or transplant patients.(35)

2.2.2 Pharmacogenomics and AI-Driven Prediction

AI has also accelerated progress in pharmacogenomics, enabling the interpretation of high-dimensional genetic data to predict drug response. Transfer learning approaches allow models trained on common variants to extrapolate predictions to rare variants of pharmacogenes such as CYP2D6 (36). Similarly, AI-driven classifiers have been applied to predict metabolizer phenotypes directly from sequencing data, bridging the gap between genomic knowledge and actionable clinical pharmacology (37) .

These developments are essential for tailoring treatment to individual patients, reducing the risks of adverse reactions, and improving therapeutic outcomes in precision medicine.

2.3 AI in Pharmacovigilance and Drug Safety

Pharmacovigilance relies on spontaneous adverse event reporting systems, which are often underreported and delayed. AI-based text mining and NLP approaches now enable real-time monitoring of EHRs, clinical notes, and social media to detect adverse drug events (ADEs).

For instance, AI systems deployed within the UK’s Yellow Card Scheme analyze millions of reports to flag potential drug safety signals earlier than manual review (38). DeepADEMiner, an NLP pipeline, mines Twitter for adverse drug effect mentions, normalizing them into standard MedDRA terminology (15)(39).

In addition, machine learning models support causality assessment, distinguishing true drug-related events from confounding factors (14). Automated pharmacovigilance not only enhances efficiency but also democratizes patient safety by including diverse data sources beyond formal clinical reports.(40)

2.4 AI in Clinical Trials and Regulatory Science

2.4.1 Clinical Trial Design and Optimization

AI has profound implications for clinical trial efficiency. Predictive modelling can optimize patient selection, sample size, and early termination criteria ((41) .For example, machine learning classifiers have been used to predict trial discontinuation risk, aiding sponsors in resource allocation (42).

SPIRIT-AI and CONSORT-AI extensions provide frameworks for designing and reporting AI-enabled interventions in clinical trials (16,17). These guidelines are critical for ensuring transparency and reproducibility of AI-assisted clinical evidence.

One of the earliest and most promising avenues for AI integration in pharmacology has been the use of electronic health record (EHR) mining. Large-scale EHR datasets, when paired with natural language processing, allow researchers to identify adverse drug events, drug–drug interactions, and off-label uses that might otherwise go unnoticed. Such approaches represent a shift from reactive to proactive pharmacovigilance, where hidden clinical patterns can be uncovered through automated text mining.(43)

Similarly, data-driven methods have been employed to predict adverse drug events before they manifest in patients. By combining prescription records with genetic and clinical information, researchers were able to anticipate unexpected drug–drug interactions, opening new perspectives for medication safety. These findings echoed earlier studies in Japanese databases, which showed that pharmacogenomic variants could be linked to adverse event frequencies in real-world prescribing.

AI has also been transformative in toxicology prediction, a critical step in drug development. Computational toxicology models, trained on structural and biochemical datasets, have achieved accuracy that surpasses traditional rule-based systems. Deep neural networks now enable early prediction of drug-induced liver injury and cardiotoxicity, reducing costly late-stage failures in pharmaceutical pipelines (44). More recently, integrative models combining transcriptomic responses and machine learning have demonstrated superior performance in forecasting compound-specific toxicities.

Together, these advances illustrate how AI not only supports clinical pharmacology but also bridges preclinical and clinical domains, creating a continuum from toxicology to bedside application. The harmonization of these approaches within regulatory frameworks will likely determine the speed at which they are embedded into pharmacological workflows.

2.4.2 Regulatory Frameworks

Regulatory agencies have acknowledged the importance of AI in pharmacology. The FDA’s database of AI/ML-enabled medical devices highlights growing acceptance, though challenges in validation remain (45). The UK’s MHRA AI roadmap (38) and the EU’s proposed Artificial Intelligence Act (46)aim to regulate AI systems according to risk levels.

In pharmacology, AI is increasingly visible in regulatory submissions for drug development, with trends showing growing integration from 2016 to 2021 (47). These initiatives underscore the dual responsibility of advancing innovation while safeguarding patient safety.

2.5 AI in Real-World Pharmacology and Medication Management

AI models are also integrated into real-world data (RWD) applications, bridging the gap between clinical trial efficacy and real-world effectiveness. RWD sources, including insurance claims, EHRs, and mobile health data, allow AI to monitor post-marketing drug outcomes, adherence, and polypharmacy risks (48).

Medication management is another domain of impact. AI-based clinical decision support systems have been deployed to reduce prescription errors and optimize therapy in chronic conditions such as anticoagulation and dialysis-related anemia (49,50). Systematic reviews indicate that AI-driven systems enhance patient safety and improve cost-effectiveness (51).

Figure 2: Traditional vs AI-Aided Drug Discovery: A Phase-by-Phase Comparison. (24)

Table 2: Applications of AI across pharmacological domains. (52)

3. ACHIEVEMENTS, CASE STUDIES, AND CURRENT APPLICATIONS OF AI IN PHARMACOLOGY

3.1 Successes in Drug Discovery and Repurposing

One of the most visible achievements of AI in pharmacology is its role in accelerating drug discovery. Traditional methods of drug design may take 10–15 years and cost billions of dollars, with a high attrition rate in late-stage trials. By contrast, AI platforms can generate candidate molecules in days, prioritize hits, and predict toxicity at early stages.

A landmark example is AlphaFold, which achieved unprecedented accuracy in protein structure prediction (26). By resolving protein conformations at near-experimental resolution, AlphaFold has transformed structure-based drug design, opening opportunities against previously inaccessible molecular targets.

Similarly, AI-driven drug repurposing provided rapid therapeutic options during the COVID-19 pandemic. The identification of baricitinib, a Janus kinase inhibitor, as a candidate treatment was facilitated by AI-based knowledge graph analysis, which highlighted its dual role in inflammation modulation and viral entry blockade (27). This case highlighted the speed and adaptability of AI, demonstrating how computational approaches can complement emergency pharmacological responses.

Other studies have shown that AI-based integrative frameworks can prioritize repurposing opportunities across Alzheimer’s disease, diabetes, and oncology (53). These approaches leverage transcriptomics, molecular networks, and drug–disease associations to discover hidden therapeutic potentials.

3.2 AI in Oncology and Personalized Medicine

Oncology has been at the forefront of AI integration. Machine learning has been employed to predict cancer susceptibility, treatment response, and prognosis. AI models trained on histopathology slides can distinguish subtle tumor features predictive of therapy outcomes (54). In breast cancer, AI systems analyzing pre-treatment tumor biopsies have successfully predicted pathological response to chemotherapy.

AI-enabled pharmacogenomics also contributes to oncology. For example, transfer learning has improved prediction of CYP2D6 haplotype functionality, enabling more precise dosing of tamoxifen and other CYP2D6-metabolized drugs (36). Beyond genetics, AI integrates multi-omics data to develop comprehensive tumor profiles guiding precision therapy.

Case studies extend to digital pathology and radiomics. Radiomic signatures derived from CT and MRI scans, analyzed via deep learning, have been used to annotate prognostic and molecular subtypes of ovarian and lung cancers (55). Such noninvasive biomarkers have potential to refine patient stratification in clinical trials.

The rise of chemoinformatics and deep learning has been a major driver of progress in drug discovery, complementing the achievements of knowledge graphs and protein structure prediction. Traditional QSAR models have now been extended through advanced neural networks, which capture non-linear and high-dimensional chemical features that were previously intractable. Similarly, transfer learning frameworks in chemistry have enabled accurate activity predictions even with limited training data, addressing a longstanding bottleneck in small-molecule discovery.

Beyond activity prediction, generative adversarial networks (GANs) have opened the door to de novo molecular design. For instance, GAN-based models have been trained to generate entirely new molecular scaffolds with desirable pharmacological properties. These approaches allow for accelerated virtual screening and prioritize candidates for wet-lab validation, cutting both costs and timelines.

At the same time, pharmacological AI has contributed to predictive toxicology in real-world contexts. Automated systems have been applied to extract drug–adverse event associations from biomedical literature and spontaneous reporting systems (15). More comprehensive reviews demonstrate that AI-driven toxicology tools can support both regulatory decision-making and clinical risk assessment (56). In addition, AI-enhanced meta-analyses of pharmacovigilance data have been shown to reduce signal detection errors compared with traditional methods (57).

Together, these advances illustrate that AI’s achievements are not limited to drug design or diagnostics but extend to the safety and sustainability of pharmacological interventions, strengthening the reliability of drug development pipelines.

3.3 Cardiovascular Pharmacology and Risk Prediction

AI has made significant progress in cardiovascular pharmacology. Large-scale studies have demonstrated that AI can predict myocardial infarction risk by integrating troponin levels with clinical variables, surpassing conventional scoring systems (58). In heart failure, machine learning models have been deployed to predict readmission and mortality, assisting clinicians in optimizing therapy (59).

Retinal fundus imaging has emerged as a surprising but powerful pharmacological biomarker source. AI models have predicted cardiovascular risk factors such as blood pressure, smoking status, and lipid levels directly from retinal images (60). Similarly, deep learning has detected anemia from fundus photographs, offering a non-invasive approach to pharmacological monitoring (61). These examples demonstrate how AI can link diagnostics to pharmacological interventions.

3.4 AI in Infectious Diseases and Pandemic Response

The COVID-19 pandemic provided a critical testbed for AI in pharmacology. Prediction models were rapidly developed to assess diagnosis, prognosis, and treatment outcomes. For example, an AI system deployed in emergency departments predicted deterioration risk among COVID-19 patients, enabling targeted pharmacological interventions (62).

Systematic reviews of AI models for COVID-19 prognosis revealed variability in quality, highlighting the need for standardized evaluation (13). Nevertheless, the pandemic demonstrated the potential of AI to accelerate drug repurposing, monitor real-world drug effects, and support adaptive trial designs under crisis conditions.

In infectious diseases beyond COVID-19, AI has been applied in antimicrobial stewardship, guiding precision dosing of antibiotics like vancomycin (63). These applications show how AI can help address antimicrobial resistance (AMR), a growing global pharmacological challenge.

3.5 AI in Neurology and Rare Diseases

AI applications extend to neurology, where drug discovery and patient monitoring are particularly challenging. In Parkinson’s disease, critical analyses have highlighted that while AI holds promise, its early evaluations have often been over-optimistic, necessitating more rigorous validation (3). Still, advances in wearable devices combined with AI signal processing allow continuous monitoring of pharmacological therapy responses, such as levodopa fluctuations.

For rare diseases, where conventional trial recruitment is difficult, AI-enabled synthetic control arms and predictive models facilitate trial feasibility. Digital biomarkers derived from imaging, electrophysiology, and omics data complement pharmacological research in this domain.

 

3.6 AI in Pharmacovigilance: Real-World Case Studies

Real-world integration of AI into pharmacovigilance systems has begun to yield results. In the UK, the MHRA implemented AI within its Yellow Card system to streamline adverse event detection and analysis, enhancing pharmacovigilance efficiency (38). In the US, FDA’s Sentinel Initiative has incorporated AI-driven analytics into safety surveillance.

Case studies show that EHR text mining identifies signals that would be missed in structured data. (64) demonstrated that free-text clinical notes could be mined with ML models to detect adverse drug reactions with high sensitivity. Additionally, social media surveillance systems, such as DeepADEMiner, have flagged real-world drug safety issues months before regulatory alerts.

These examples illustrate the shift from passive, retrospective safety monitoring to proactive, predictive pharmacovigilance supported by AI.

3.7 Clinical Decision Support and Medication Management

Clinical decision support (CDS) systems powered by AI are increasingly deployed in hospitals and pharmacies. In dialysis patients, AI-based CDS improved anemia management outcomes while optimizing erythropoietin dosing (50). Similarly, AI-guided anticoagulation management reduced the risk of nonadherence and thromboembolic events (49). Systematic reviews confirm that AI-based interventions enhance medication safety by reducing prescription errors, alert fatigue, and drug–drug interactions (51,65). These real-world achievements demonstrate the scalability of AI across diverse pharmacological contexts.

Figure 3: Core Applications of Artificial Intelligence in Pharmacology and Pharmaceutical Sciences.

Table 3: Core achievements and applications of AI in pharmacology

Area

Representative AI methods

Key impact

Drug discovery (structure)

Deep learning for protein structure (AlphaFold)

Unlocks hard targets; speeds rational design

Drug repurposing

Knowledge graphs; multi-omics networks

Rapid candidate ID (e.g., COVID baricitinib); cross-disease prioritization

Oncology & precision medicine

Histopathology DL; pharmacogenomics (transfer learning); radiomics

Better prognosis/response prediction; dosing refinement; trial stratification

Cardiovascular risk

ML on troponin + clinical vars; fundus DL

Outperforms scores; noninvasive CV/anemia screening

Infectious diseases

ED deterioration DL; ML dosing (vancomycin); evidence appraisals

Targeted interventions; stewardship; calls for robust standards

Pharmacovigilance

EHR NLP; national systems; social data mining

Earlier ADR signals; faster triage

Clinical decision support

AI-CDS (dosing/management)

Improved outcomes in dialysis anemia; safer anticoagulation

Predictive toxicology

NLP/ML on literature & PV; DL-tox

Earlier toxicity detection; supports regulatory review

4. CHALLENGES, RISKS, AND LIMITATIONS OF AI IN PHARMACOLOGY

4.1 Technical Challenges

4.1.1 Data Quality and Availability

AI in pharmacology depends on access to large, diverse, and high-quality datasets. However, biomedical and pharmacological data are often fragmented, incomplete, biased, or unstandardized. Electronic health records (EHRs) frequently contain missing or inconsistent values, which can mislead algorithms unless carefully addressed through imputation or robust model design (10,66). Furthermore, pharmacovigilance databases such as the FDA’s FAERS suffer from under-reporting and lack of denominator data, limiting their utility without careful preprocessing.

4.1.2 Hidden Stratification and Dataset Shift

Hidden stratification occurs when subgroups within datasets are not adequately represented or labeled, leading to models that perform well in aggregate but fail in specific subpopulations. This issue is particularly critical in pharmacology, where genetic variability, comorbidities, and age can strongly influence drug response (67). Dataset shift, in which the distribution of data changes between training and deployment environments, also undermines reliability (68)

4.1.3 Reproducibility and Transparency

Many AI models in pharmacology remain black-box systems, raising concerns about reproducibility and interpretability. Studies often report high accuracy on internal datasets but fail when tested externally. Without transparent methodologies, including open-source code, clear reporting (CONSORT-AI, SPIRIT-AI), and independent validation, translation into real-world pharmacology remains uncertain (16,17).

4.1.4 Explainability

Explainable AI (XAI) is essential for clinical adoption. Pharmacologists and clinicians need to understand why a model makes a recommendation, especially when it involves critical decisions like drug dosing or safety alerts. Techniques such as saliency maps and attention mechanisms provide some interpretability, but remain imperfect (69). The gap between model performance and explainability remains a persistent barrier.

A further set of challenges relates to the safety and reliability of pharmacological AI systems when deployed in real-world practice. Early investigations demonstrated that even well-calibrated machine learning models can fail to generalize across populations, leading to unexpected clinical outcomes. More recent work using large-scale pharmacovigilance databases has highlighted how machine learning can both improve and complicate adverse drug reaction detection, depending on data quality and feature representation.

Another major barrier lies in the under-reporting of adverse drug reactions (ADRs), a well-documented problem that limits the robustness of any AI model trained on pharmacovigilance data. Without strong reporting incentives, algorithms risk perpetuating existing blind spots in drug safety.

To address this, integrative data resources have been developed that link chemical, biological, and clinical domains. Databases such as DrugBank and STITCH offer curated connections between compounds, targets, and ADRs, serving as valuable foundations for AI-based pharmacological models. However, concerns remain regarding data standardization and the reproducibility of findings across platforms.

Finally, in low- and middle-income countries, AI pharmacovigilance must confront infrastructural and regulatory hurdles. Studies emphasize the importance of local adaptation, as systems trained on Western data may misrepresent risks in Asian or African contexts. Unless datasets are inclusive and context-aware, AI will risk reinforcing global health inequities rather than alleviating them.

4.2 Ethical, Legal, and Social Challenges

4.2.1 Algorithmic Bias

Bias in AI systems arises from unrepresentative datasets or flawed design. In pharmacology, biased models can amplify health inequities, leading to inappropriate dosing, under-detection of adverse events, or exclusion of vulnerable populations (70,71). For example, if genomic datasets underrepresent minority populations, pharmacogenomic predictions may be skewed.

4.2.2 Accountability and Liability

Determining legal responsibility for AI-driven pharmacological decisions remains unresolved. If an AI-based dosing system leads to an adverse event, should liability rest with the clinician, the hospital, the software vendor, or the regulator? Legal scholars emphasize the need for clear liability frameworks before AI can be safely embedded in pharmacology (72).

4.2.3 Privacy and Security

Pharmacological AI often requires access to sensitive patient-level data. This raises issues of data protection, consent, and cybersecurity. Breaches of medical AI systems could expose genetic or prescription data, eroding trust in pharmacology research. Emerging solutions include federated learning (training models across distributed datasets without centralizing data), though implementation remains complex.

4.3 Systemic and Regulatory Challenges

4.3.1 Regulatory Hesitancy

Regulators are still adapting to AI’s unique challenges. Unlike conventional medical devices or drugs, AI algorithms may evolve over time (continuous learning systems), making static regulatory approval insufficient (45,73). While agencies like the FDA and EMA have begun issuing guidance, frameworks remain fragmented, especially for pharmacological applications such as adaptive dosing or trial optimization.

4.3.2 Lack of Standardized Frameworks

Although reporting standards such as SPIRIT-AI and CONSORT-AI exist, their adoption in pharmacology research is inconsistent (16,17). Without mandatory compliance, reproducibility and trust remain limited.

4.3.3 Cost-Effectiveness and Scalability

AI solutions may be costly to develop and deploy. Smaller pharmaceutical companies and public-sector pharmacology departments may struggle to access the computational infrastructure required for advanced AI. Even when effective, AI systems must be scaled and integrated into existing workflows, which is often more complex than initial pilot studies suggest.

4.4 Societal Concerns and Clinical Trust

4.4.1 Human–AI Collaboration

Clinicians and pharmacologists may be reluctant to trust AI-generated recommendations, particularly when they lack transparency or appear to contradict clinical experience. Studies reveal that trust is built when AI tools are framed as assistive rather than replacement technologies (19). This concept of “augmented intelligence” emphasizes human oversight, maintaining clinical responsibility while benefiting from AI insights.

4.4.2 Risk of Widening Inequalities

AI has the potential to worsen global inequities in pharmacology. High-income countries with advanced computational infrastructure may accelerate AI-driven discoveries, while low- and middle-income countries (LMICs) risk being left behind. Moreover, datasets predominantly sourced from Western populations may not generalize globally, further exacerbating disparities (74).

4.5 Case Studies Illustrating Limitations

  • COVID-19 Prediction Models: Early AI models predicting COVID-19 prognosis performed poorly when externally validated, reflecting overfitting and lack of generalizability (13).
  • Hidden Bias in Imaging: Models trained on imaging datasets have failed in real-world deployments due to hidden stratification, leading to clinically meaningful errors (67).
  • Drug Safety Monitoring: Despite advances, pharmacovigilance systems still suffer from false positives when mining social media data, requiring careful human oversight (56).

These examples emphasize that while AI shows promise, it must be rigorously validated and integrated cautiously.

4.6 Potential Solutions to Current Challenges

Efforts are underway to address these limitations. Federated learning and privacy-preserving AI may reduce data-sharing risks. Explainable AI techniques are advancing toward more interpretable decision support. Regulatory sandboxes, where AI tools are tested in controlled environments, offer a safe space for iterative improvement. Finally, cross-disciplinary education of pharmacologists, clinicians, and data scientists can improve mutual understanding and trust.

Table 4: Core challenges and pragmatic mitigations in AI-driven pharmacology

Key challenge (summary)

Main risk

Practical mitigation

Data quality & availability (fragmented EHR/PV data; under-reporting)

Spurious models; unsafe recs

Data standards + rigorous curation; federated/PPML; multi-site external validation

Hidden stratification & dataset shift

Inequitable/unstable performance

Subgroup audits; continuous drift monitoring; diverse training data

Reproducibility & explainability (black boxes)

Low trust; poor generalization

SPIRIT-AI/CONSORT-AI; transparent methods; intrinsic/post-hoc XAI

Algorithmic bias, accountability, privacy

Inequitable dosing; legal uncertainty; breaches

Equity-by-design; defined liability/governance; access control + federated learning

Regulatory hesitancy, lack of standards, scalability

Stalled adoption; inconsistent evidence

Regulatory sandboxes; enforce reporting standards; realistic costed deployment plans

Real-world failures (COVID models, imaging bias, noisy PV)

Harmful/incorrect actions

Prospective external validation; human-in-the-loop adjudication

5. FUTURE PROSPECTS, ROADMAP, AND CONCLUSION

5.1 Blockchain and AI in Pharmacology

One of the most promising intersections is the combination of blockchain technology and AI. Pharmacology depends on secure, transparent, and traceable data, particularly in clinical trials, drug supply chains, and pharmacovigilance. Blockchain, with its decentralized and tamper-proof ledger, can ensure integrity and transparency, while AI provides the analytic power to interpret and act on these data streams.

For instance, clinical trial registries on blockchain could prevent selective reporting or data manipulation, ensuring accountability across sponsors and regulators (75). When integrated with AI, such systems could automatically analyze trial data, flag anomalies, and optimize trial endpoints. Similarly, blockchain-secured supply chains can prevent counterfeit drugs, a major pharmacological and public health threat in low- and middle-income countries.

In pharmacovigilance, blockchain-enabled patient reporting systems could empower individuals to report adverse events directly, with AI aggregating and analyzing these inputs in real time. This dual system could revolutionize post-marketing surveillance by enhancing both security and efficiency.

Figure 5: Integration of Blockchain and AI in Drug Discovery & Healthcare Systems. Made with Mermaid Website

5.2 Federated Learning and Privacy-Preserving AI

A major barrier to AI adoption in pharmacology is the sensitivity of patient-level data. Federated learning offers a solution by enabling AI models to be trained across multiple institutions without centralizing data. This method preserves privacy while expanding the diversity of training data, reducing algorithmic bias (71).

In pharmacogenomics, federated learning could pool sequencing data from global populations while keeping sensitive information local. In pharmacovigilance, multi-country data could be analyzed collaboratively to identify safety signals earlier, without violating jurisdictional privacy laws such as GDPR in the EU or HIPAA in the US.

Edge AI, which deploys models directly on devices such as hospital servers or mobile phones, complements this trend by minimizing dependence on cloud infrastructure. Together, federated and edge AI point toward a future where pharmacological insights are derived from diverse, global datasets without compromising security.

5.3 Digital Twins in Pharmacology

The concept of digital twins—virtual models of individual patients—has attracted significant attention in medicine and pharmacology. By integrating clinical data, genomics, pharmacokinetics, and environmental variables, a digital twin can simulate drug responses and predict outcomes before real-world administration (76).

In oncology, digital twins could simulate tumor growth and test different chemotherapy regimens in silico, guiding personalized treatment. In clinical pharmacology, twins could help optimize dosing in populations with high variability, such as pediatrics or geriatrics. Moreover, they can serve as synthetic control arms in clinical trials, reducing the burden of recruitment for rare diseases.

Although the concept is still in early stages, pilot projects demonstrate feasibility. Integrating AI-driven pharmacological models with real-world feedback loops could make digital twins an integral tool for precision medicine in the coming decades.

5.4 Multi-Omics Integration and Systems Pharmacology

Pharmacology is increasingly moving from reductionist models toward systems-level approaches, where multiple layers of biological data are integrated. AI is uniquely positioned to handle the complexity of multi-omics data—genomics, transcriptomics, proteomics, metabolomics, and microbiomics—linking them with pharmacokinetics and pharmacodynamics (PK/PD).

For example, deep learning models can correlate microbiome profiles with drug metabolism variability, providing insights into why certain patients exhibit poor response or severe toxicity. Similarly, integrating proteomic and metabolomic data enhances biomarker discovery for drug efficacy prediction.

AI-enabled systems pharmacology can also support drug repurposing by mapping entire biological networks, identifying nodes where existing drugs may have unrecognized therapeutic effects. This holistic approach is expected to dominate the next generation of pharmacological research.

One of the most promising future directions lies in the integration of AI with drug discovery and repurposing pipelines. Traditional drug development remains time-consuming and costly, but AI-based models are increasingly able to identify novel targets, predict molecular interactions, and even design new compounds in silico. Notably, deep learning–driven QSAR models have demonstrated the ability to handle high-dimensional chemical descriptor data, leading to more accurate toxicity prediction and early candidate prioritization (23)

Recent research has also emphasized drug repurposing, where AI algorithms mine diverse biological and clinical datasets to suggest new indications for existing compounds. Notable case studies include Alzheimer’s disease, where AI frameworks identified new candidate molecules for repositioning (53), and oncology, where systematic integration of biomedical knowledge helped prioritize clinically relevant drugs (28).

Furthermore, generative adversarial networks (GANs) and advanced neural architectures have emerged as powerful tools for creating novel molecular structures with desirable pharmacokinetic and pharmacodynamic properties. Such approaches complement precision medicine initiatives, allowing personalized drug design aligned with patient-specific genetic and metabolic profiles.

While the potential is vast, these advances also necessitate careful regulatory oversight and validation before clinical adoption. However, the convergence of AI, cheminformatics, and computational biology is widely regarded as one of the most transformative frontiers in pharmacology, bridging discovery with real-world clinical application.

5.5 Global Health, Policy, and Education

5.5.1 Policy and Regulation

Policymakers are actively shaping the regulatory landscape for AI in pharmacology. The European Union’s AI Act (2021) (46,77) categorizes AI systems by risk level, imposing stricter requirements on high-risk applications such as medical dosing algorithms. Similarly, the FDA has launched a regulatory framework for adaptive AI/ML-based software ((45,73). These initiatives highlight the importance of balancing innovation with patient safety. In India, the NITI Aayog has launched the National Strategy for Artificial Intelligence, emphasizing applications in healthcare, including pharmacology. The UK’s NHS AI Lab also funds projects aimed at integrating AI into clinical workflows. Globally, the World Health Organization (WHO) has published guidance emphasizing equity, transparency, and accountability.

5.5.2 Education and Workforce Training

Integrating AI into pharmacology requires a new skill set for pharmacologists, clinicians, and regulators. Training programs must include bioinformatics, data science, and AI ethics alongside classical pharmacology. Multidisciplinary collaboration is essential: pharmacologists provide clinical expertise, data scientists develop algorithms, and ethicists ensure responsible deployment.

5.5.3 Equity and Access

AI must not exacerbate global health inequities. Ensuring that LMICs have access to AI-enabled pharmacology, through open-access datasets, capacity building, and affordable infrastructure, is a moral and scientific imperative. International collaborations can ensure that the benefits of AI are equitably distributed across populations.

5.6 Roadmap for the Future

The integration of AI into pharmacology requires a structured roadmap:

  1. Standardization: Develop harmonized data formats, reporting guidelines (SPIRIT-AI, CONSORT-AI), and benchmarking datasets.
  2. Validation: Ensure rigorous external validation of models, ideally across multiple healthcare systems and populations.
  3. Transparency: Promote explainable AI to build trust among clinicians and pharmacologists.
  4. Collaboration: Foster interdisciplinary teams linking pharmacology, computer science, regulatory science, and ethics.
  5. Scalability: Move from pilot projects to scalable, sustainable implementations in clinical and research environments.
  6. Global Perspective: Encourage cross-border collaborations and international regulatory harmonization to enable global AI pharmacology.

CONCLUSION

Artificial intelligence has moved from theoretical promise to practical reality in pharmacology. It has accelerated drug discovery, supported precision dosing, enhanced pharmacovigilance, and optimized clinical trial design. Achievements such as AlphaFold, AI-guided repurposing of baricitinib, and AI-enhanced pharmacogenomics highlight the breadth of progress to date. Yet, challenges remain. Issues of data quality, bias, interpretability, and regulation must be addressed before AI can be fully integrated into everyday pharmacology. Ethical considerations—privacy, accountability, equity—are as critical as technical innovation. Looking forward, AI will likely become the foundation of systems pharmacology, powered by blockchain, federated learning, digital twins, and multi-omics integration. These innovations hold the potential not only to transform pharmacological science but also to deliver tangible benefits to patients worldwide. The vision is clear: a future in which AI augments pharmacology, enabling safer, faster, and more personalized therapies.

REFERENCES

  1. Badillo S, Banfai B, Birzele F, et al. An introduction to machine learning. Clin Pharmacol Ther. 2020;107(4):871–85.
  2. Ross J, Webb C, Rahman F. Artificial Intelligence in Healthcare. 2023.
  3. Ge W, Lueck C, Suominen H, Apthorp D. Has machine learning over-promised in healthcare? A critical analysis and a proposal for improved evaluation, with evidence from Parkinson’s disease. Artif Intell Med. 2023;139:102524.
  4. Meskó B, Görög M. A short guide for medical professionals in the era of artificial intelligence. NPJ Digit Med. 2020;3:126.
  5. Mahesh B. Machine learning algorithms – a review. Int J Sci Res. 2020;9(1):381–6.
  6. Xiao C, Choi E, Sun J. Opportunities and challenges in developing deep learning models using electronic health records data: a systematic review. Journal of the American Medical Informatics Association. 2018;25(10):1419–28.
  7. Choi RY, Coyner AS, Kalpathy-Cramer J, Chiang MF, Campbell JP. Introduction to machine learning, neural networks, and deep learning. Transl Vis Sci Technol. 2020;9:14.
  8. Rajkomar A, Dean J, Kohane IS. Machine learning in medicine. New England Journal of Medicine. 2019;380(14):1347–58.
  9. Vaswani A, Shazeer N, Parmar N, et al. Attention Is All You Need. In: Advances in Neural Information Processing Systems. Curran Associates, Inc.; 2017.
  10. Yoon J, Jordon J, van der Schaar M. GAIN: Missing data imputation using generative adversarial nets. In: Proceedings of the 35th International Conference on Machine Learning. PMLR; 2018. p. 5689–98.
  11. Ribba B, Dudal S, Lavé T, Peck RW. Model-informed artificial intelligence: reinforcement learning for precision dosing. Clin Pharmacol Ther. 2020;107:853–7.
  12. Gupta R, Kurc T, Sharma A, Almeida JS, Saltz J. The emergence of Pathomics. Curr Pathobiol Rep. 2019;7:73–84.
  13. Wynants L, Van Calster B, Collins GS, et al. Prediction models for diagnosis and prognosis of COVID-19: systematic review and critical appraisal. BMJ. 2020;369:m1328.
  14. Cherkas Y, Ide J, van Stekelenborg J. Leveraging machine learning to facilitate individual case causality assessment of adverse drug reactions. Drug Saf. 2022;45(5):571–82.
  15. Sarker A, Ginn R, Nikfarjam A, et al. Utilizing social media data for pharmacovigilance: a review. J Biomed Inform. 2015;54:202–12.
  16. Cruz Rivera S, Liu X, Chan AW, et al. Guidelines for clinical trial protocols for interventions involving artificial intelligence: the SPIRIT-AI extension. Nat Med. 2020;26(9):1351–63.
  17. Liu X, Cruz Rivera S, Moher D, Calvert MJ, Denniston AK. Reporting guidelines for clinical trial reports for interventions involving artificial intelligence: the CONSORT-AI extension. Nat Med. 2020;26:1364–74.
  18. Mayr A, Klambauer G, Unterthiner T, Hochreiter S. DeepTox: toxicity prediction using deep learning. Front Environ Sci. 2016;3:80.
  19. World Medical Association. WMA statement on augmented intelligence in medical care. 2022.
  20. Patel VL, Kannampallil TG. Cognitive informatics in biomedicine and healthcare. J Biomed Inform. 2015;53:3–14.
  21. Miotto R, Wang F, Wang S, Jiang X, Dudley JT. Deep learning for healthcare: review, opportunities and challenges. Brief Bioinform. 2017;19(6):1236–46.
  22. Corrigan BW. Artificial intelligence and machine learning: will clinical pharmacologists be needed in the next decade? The John Henry question. Clin Pharmacol Ther. 2020;107(4):697–9.
  23. Roy K, Kar S, Das RN. Understanding the Basics of QSAR for Applications in Pharmaceutical Sciences and Risk Assessment. Academic Press; 2015.
  24. Jarallah SJ, Almughem FA, Alhumaid NK, Fayez N AL, Alradwan I, Alsulami KA, et al. Artificial intelligence revolution in drug discovery: A paradigm shift in pharmaceutical innovation. Int J Pharm. 2025 Jul;680:125789.
  25. Karimi M, Wu D, Wang Z, Shen Y. DeepAffinity: Interpretable deep learning of compound-protein affinity. Bioinformatics. 2019;35(18):3329–38.
  26. Jumper J, Evans R, Pritzel A, et al. Highly accurate protein structure prediction with AlphaFold. Nature. 2021;596(7873):583–9.
  27. Richardson P, Griffin I, Tucker C, et al. Baricitinib as potential treatment for 2019-nCoV acute respiratory disease. The Lancet. 2020;395(10223):e30–e31.
  28. Himmelstein DS, Lizee A, Hessler C, et al. Systematic integration of biomedical knowledge prioritizes drugs for repurposing. Elife. 2017;6:e26726.
  29. Williams DP, Lazic SE, Foster AJ, Semenova E, Morgan P. Predicting drug-induced liver injury with Bayesian machine learning. Chem Res Toxicol. 2020;33:239–48.
  30. Al-Taie Z, Liu D, Mitchem JB, et al. Explainable AI in high-throughput drug repositioning for subgroup stratifications. J Biomed Inform. 2021;118:103792.
  31. Semenova E, Williams DP, Afzal AM, Lazic SE. A Bayesian neural network for toxicity prediction. Computational Toxicology. 2020;16:100133.
  32. Woillard JB, Labriffe M, Debord J, Marquet P. Mycophenolic acid exposure prediction using machine learning. Clin Pharmacol Ther. 2021;110:370–9.
  33. Woillard JB, Labriffe M, Prémaud A, Marquet P. Estimation of drug exposure by ML based on simulations from published PK models: tacrolimus example. Pharmacology Research. 2021;167:105578.
  34. Destere A, Marquet P, Labriffe M, Drici MD, Woillard JB. A hybrid algorithm combining population pharmacokinetic and machine learning for isavuconazole exposure prediction. Pharm Res. 2023;40(4):951–9.
  35. van Gelder T, Vinks AA. Machine learning as a novel method to support therapeutic drug management and precision dosing. Clin Pharmacol Ther. 2021;110:273–6.
  36. McInnes G, Dalton R, Sangkuhl K, et al. Transfer learning enables prediction of CYP2D6 haplotype function. PLoS Comput Biol. 2020;16:e1008399.
  37. van der Lee M, Allard WG, Vossen RHAM, et al. Toward predicting CYP2D6-mediated variable drug response from gene sequencing data. Sci Transl Med. 2021;13(603):eabf3637.
  38. UK MHRA. Software and AI as a medical device change programme—roadmap. 2024.
  39. DeepADEMiner: a deep learning pharmacovigilance pipeline for extraction and normalization of adverse drug effect mentions on Twitter. 2020.
  40. Dev S, Zhang S, Voyles J, Rao AS. Automated classification of adverse events in pharmacovigilance. Proceedings of IEEE BIBM. 2017;1562–6.
  41. Harrer S, Shah P, Antony B, Hu J. Artificial intelligence for clinical trial design. Trends Pharmacol Sci. 2019;40(8):577–91.
  42. Kavalci E, Hartshorn A. Improving clinical trial design using interpretable machine learning based prediction of early trial termination. Sci Rep. 2023;13:121.
  43. Luo Y, Thompson WK, Herr TM, Zeng Z, Berendsen MA, Jonnalagadda SR, et al. Natural language processing for EHR-based pharmacovigilance: a structured review. Drug Saf. 2017;40(11):1075–89.
  44. Mamoshina P, Bueno-Orovio A, Rodriguez B. Dual transcriptomic and molecular machine learning predicts all major clinical forms of drug cardiotoxicity. Front Pharmacol. 2020;11:639.
  45. Ebrahimian S, Kalra MK, Agarwal S, et al. FDA-regulated AI algorithms: trends, strengths, and gaps of validation studies. Acad Radiol. 2022;29(4):559–66.
  46. European Commission. Proposal for a Regulation laying down harmonised rules on artificial intelligence (Artificial Intelligence Act). 2021.
  47. Liu Q, Huang R, Hsieh J, et al. Landscape analysis of the application of artificial intelligence and machine learning in regulatory submissions for drug development from 2016 to 2021. Clin Pharmacol Ther. 2023;113(4):771–4.
  48. Liu F, Panagiotakos D. Real-world data: methods, applications, challenges and opportunities. BMC Med Res Methodol. 2022;22:287.
  49. Labovitz DL, Shafner L, Reyes Gil M, Virmani D, Hanina A. Using artificial intelligence to reduce the risk of nonadherence in patients on anticoagulation therapy. Stroke. 2017;48(5):1416–9.
  50. Barbieri C, Molina M, Ponce P, et al. AI clinical decision support optimizes anemia management in hemodialysis patients. Kidney Int. 2016;90(2):422–9.
  51. Damiani G, Altamura G, Zedda M, et al. Potentiality of algorithms and AI adoption to improve medication management in primary care: a systematic review. BMJ Open. 2023;13:e065301.
  52. Paul D, Sanap G, Shenoy S, Kalyane D, Kalia K, Tekade RK. Artificial intelligence in drug discovery and development. Drug Discov Today. 2021 Jan;26(1):80–93.
  53. Fang J, Zhang P, Wang Q, et al. AI framework identifies candidate targets for drug repurposing in Alzheimer’s disease. Alzheimers Res Ther. 2022;14:7.
  54. Saednia K, Lagree A, Alera MA, et al. Quantitative digital histopathology and machine learning to predict pathological complete response to chemotherapy in breast cancer. Sci Rep. 2022;12(1):9690.
  55. Lu H, Arshad M, Thornton A, et al. A mathematical descriptor of tumor mesoscopic structure from CT images annotates prognostic and molecular phenotypes of epithelial ovarian cancer. Nat Commun. 2019;10:764.
  56. Tricco AC, Zarin W, Lillie E, et al. Utility of social media and crowd-intelligence data for pharmacovigilance: a scoping review. BMC Med Inform Decis Mak. 2018;18(1):38.
  57. Comfort S, Perera S, Hudson Z, et al. Sorting through the safety data haystack: using ML to identify ICSRs in social digital media. Drug Saf. 2018;41(6):579–90.
  58. Doudesis D, Lee KK, Boeddinghaus J, et al. Machine learning for diagnosis of myocardial infarction using cardiac troponin concentrations. Nat Med. 2023;29(5):1201–10.
  59. Guo A, Pasque M, Loh F, Mann DL, Payne PRO. Heart failure diagnosis, readmission, and mortality prediction using machine learning and AI models. Curr Epidemiol Rep. 2020;7(4):212–9.
  60. Poplin R, Varadarajan A V, Blumer K, et al. Prediction of cardiovascular risk factors from retinal fundus photographs via deep learning. Nat Biomed Eng. 2018;2(3):158–64.
  61. Mitani A, Huang A, Venugopalan S, et al. Detection of anaemia from retinal fundus images via deep learning. Nat Biomed Eng. 2020;4(1):18–27.
  62. Shamout FE, Shen Y, Wu N, et al. An artificial intelligence system for predicting the deterioration of COVID-19 patients in the emergency department. NPJ Digit Med. 2021;4:1–11.
  63. Wang Z, Ong CLJ, Fu Z. AI models to assist vancomycin dosage titration. Front Pharmacol. 2022;13:801928.
  64. Wasylewicz A, van de Burgt B, Weterings A, et al. Identifying adverse drug reactions from free-text electronic hospital health record notes. Br J Clin Pharmacol. 2022;88(3):1235–45.
  65. Rozenblum R, Rodriguez-Monguio R, Volk LA, et al. Using a machine learning system to identify and prevent medication prescribing errors: a clinical and cost analysis evaluation. Jt Comm J Qual Patient Saf. 2020;46(1):3–10.
  66. Shah AD, Bartlett JW, Carpenter J, Nicholas O, Hemingway H. Comparison of random forest and parametric imputation models for imputing missing data using MICE: a CALIBER study. Am J Epidemiol. 2014;179(6):764–74.
  67. Oakden-Rayner L, Dunnmon J, Carneiro G, Ré C. Hidden stratification causes clinically meaningful failures in ML for medical imaging. 2019.
  68. Finlayson SG, Subbaswamy A, Singh K, et al. The clinician and dataset shift in artificial intelligence. New England Journal of Medicine. 2021;385(3):283–6.
  69. Mundhenk TN, Chen BY, Friedland G. Efficient saliency maps for explainable AI. 2020.
  70. Vokinger KN, Feuerriegel S, Kesselheim AS. Mitigating bias in machine learning for medicine. Communications Medicine. 2021;1:1–3.
  71. Panch T, Mattie H, Atun R. Artificial intelligence and algorithmic bias: implications for health systems. J Glob Health. 2019;9:20318.
  72. Price WN, Cohen IG. Locating liability for medical AI. 2023.
  73. FDA CDRH. Artificial intelligence and machine learning in software as a medical device. 2022.
  74. Veinot TC, Mitchell H, Ancker JS. Good intentions are not enough: how informatics interventions can worsen inequality. Journal of the American Medical Informatics Association. 2018;25(8):1080–8.
  75. Kumar Y, Gupta S, Singla R, Hu YC. A systematic review of artificial intelligence techniques in cancer prediction and diagnosis. Archives of Computational Methods in Engineering. 2022;29(4):2043–70.
  76. Zhavoronkov A, Vanhaelen Q, Oprea TI. Will artificial intelligence for drug discovery impact clinical pharmacology? Clin Pharmacol Ther. 2020;107(4):780–5.
  77. European Parliament Research Services. Artificial Intelligence in Healthcare: Applications, Risks, and Ethical and Societal Impacts. 2022.

Reference

  1. Badillo S, Banfai B, Birzele F, et al. An introduction to machine learning. Clin Pharmacol Ther. 2020;107(4):871–85.
  2. Ross J, Webb C, Rahman F. Artificial Intelligence in Healthcare. 2023.
  3. Ge W, Lueck C, Suominen H, Apthorp D. Has machine learning over-promised in healthcare? A critical analysis and a proposal for improved evaluation, with evidence from Parkinson’s disease. Artif Intell Med. 2023;139:102524.
  4. Meskó B, Görög M. A short guide for medical professionals in the era of artificial intelligence. NPJ Digit Med. 2020;3:126.
  5. Mahesh B. Machine learning algorithms – a review. Int J Sci Res. 2020;9(1):381–6.
  6. Xiao C, Choi E, Sun J. Opportunities and challenges in developing deep learning models using electronic health records data: a systematic review. Journal of the American Medical Informatics Association. 2018;25(10):1419–28.
  7. Choi RY, Coyner AS, Kalpathy-Cramer J, Chiang MF, Campbell JP. Introduction to machine learning, neural networks, and deep learning. Transl Vis Sci Technol. 2020;9:14.
  8. Rajkomar A, Dean J, Kohane IS. Machine learning in medicine. New England Journal of Medicine. 2019;380(14):1347–58.
  9. Vaswani A, Shazeer N, Parmar N, et al. Attention Is All You Need. In: Advances in Neural Information Processing Systems. Curran Associates, Inc.; 2017.
  10. Yoon J, Jordon J, van der Schaar M. GAIN: Missing data imputation using generative adversarial nets. In: Proceedings of the 35th International Conference on Machine Learning. PMLR; 2018. p. 5689–98.
  11. Ribba B, Dudal S, Lavé T, Peck RW. Model-informed artificial intelligence: reinforcement learning for precision dosing. Clin Pharmacol Ther. 2020;107:853–7.
  12. Gupta R, Kurc T, Sharma A, Almeida JS, Saltz J. The emergence of Pathomics. Curr Pathobiol Rep. 2019;7:73–84.
  13. Wynants L, Van Calster B, Collins GS, et al. Prediction models for diagnosis and prognosis of COVID-19: systematic review and critical appraisal. BMJ. 2020;369:m1328.
  14. Cherkas Y, Ide J, van Stekelenborg J. Leveraging machine learning to facilitate individual case causality assessment of adverse drug reactions. Drug Saf. 2022;45(5):571–82.
  15. Sarker A, Ginn R, Nikfarjam A, et al. Utilizing social media data for pharmacovigilance: a review. J Biomed Inform. 2015;54:202–12.
  16. Cruz Rivera S, Liu X, Chan AW, et al. Guidelines for clinical trial protocols for interventions involving artificial intelligence: the SPIRIT-AI extension. Nat Med. 2020;26(9):1351–63.
  17. Liu X, Cruz Rivera S, Moher D, Calvert MJ, Denniston AK. Reporting guidelines for clinical trial reports for interventions involving artificial intelligence: the CONSORT-AI extension. Nat Med. 2020;26:1364–74.
  18. Mayr A, Klambauer G, Unterthiner T, Hochreiter S. DeepTox: toxicity prediction using deep learning. Front Environ Sci. 2016;3:80.
  19. World Medical Association. WMA statement on augmented intelligence in medical care. 2022.
  20. Patel VL, Kannampallil TG. Cognitive informatics in biomedicine and healthcare. J Biomed Inform. 2015;53:3–14.
  21. Miotto R, Wang F, Wang S, Jiang X, Dudley JT. Deep learning for healthcare: review, opportunities and challenges. Brief Bioinform. 2017;19(6):1236–46.
  22. Corrigan BW. Artificial intelligence and machine learning: will clinical pharmacologists be needed in the next decade? The John Henry question. Clin Pharmacol Ther. 2020;107(4):697–9.
  23. Roy K, Kar S, Das RN. Understanding the Basics of QSAR for Applications in Pharmaceutical Sciences and Risk Assessment. Academic Press; 2015.
  24. Jarallah SJ, Almughem FA, Alhumaid NK, Fayez N AL, Alradwan I, Alsulami KA, et al. Artificial intelligence revolution in drug discovery: A paradigm shift in pharmaceutical innovation. Int J Pharm. 2025 Jul;680:125789.
  25. Karimi M, Wu D, Wang Z, Shen Y. DeepAffinity: Interpretable deep learning of compound-protein affinity. Bioinformatics. 2019;35(18):3329–38.
  26. Jumper J, Evans R, Pritzel A, et al. Highly accurate protein structure prediction with AlphaFold. Nature. 2021;596(7873):583–9.
  27. Richardson P, Griffin I, Tucker C, et al. Baricitinib as potential treatment for 2019-nCoV acute respiratory disease. The Lancet. 2020;395(10223):e30–e31.
  28. Himmelstein DS, Lizee A, Hessler C, et al. Systematic integration of biomedical knowledge prioritizes drugs for repurposing. Elife. 2017;6:e26726.
  29. Williams DP, Lazic SE, Foster AJ, Semenova E, Morgan P. Predicting drug-induced liver injury with Bayesian machine learning. Chem Res Toxicol. 2020;33:239–48.
  30. Al-Taie Z, Liu D, Mitchem JB, et al. Explainable AI in high-throughput drug repositioning for subgroup stratifications. J Biomed Inform. 2021;118:103792.
  31. Semenova E, Williams DP, Afzal AM, Lazic SE. A Bayesian neural network for toxicity prediction. Computational Toxicology. 2020;16:100133.
  32. Woillard JB, Labriffe M, Debord J, Marquet P. Mycophenolic acid exposure prediction using machine learning. Clin Pharmacol Ther. 2021;110:370–9.
  33. Woillard JB, Labriffe M, Prémaud A, Marquet P. Estimation of drug exposure by ML based on simulations from published PK models: tacrolimus example. Pharmacology Research. 2021;167:105578.
  34. Destere A, Marquet P, Labriffe M, Drici MD, Woillard JB. A hybrid algorithm combining population pharmacokinetic and machine learning for isavuconazole exposure prediction. Pharm Res. 2023;40(4):951–9.
  35. van Gelder T, Vinks AA. Machine learning as a novel method to support therapeutic drug management and precision dosing. Clin Pharmacol Ther. 2021;110:273–6.
  36. McInnes G, Dalton R, Sangkuhl K, et al. Transfer learning enables prediction of CYP2D6 haplotype function. PLoS Comput Biol. 2020;16:e1008399.
  37. van der Lee M, Allard WG, Vossen RHAM, et al. Toward predicting CYP2D6-mediated variable drug response from gene sequencing data. Sci Transl Med. 2021;13(603):eabf3637.
  38. UK MHRA. Software and AI as a medical device change programme—roadmap. 2024.
  39. DeepADEMiner: a deep learning pharmacovigilance pipeline for extraction and normalization of adverse drug effect mentions on Twitter. 2020.
  40. Dev S, Zhang S, Voyles J, Rao AS. Automated classification of adverse events in pharmacovigilance. Proceedings of IEEE BIBM. 2017;1562–6.
  41. Harrer S, Shah P, Antony B, Hu J. Artificial intelligence for clinical trial design. Trends Pharmacol Sci. 2019;40(8):577–91.
  42. Kavalci E, Hartshorn A. Improving clinical trial design using interpretable machine learning based prediction of early trial termination. Sci Rep. 2023;13:121.
  43. Luo Y, Thompson WK, Herr TM, Zeng Z, Berendsen MA, Jonnalagadda SR, et al. Natural language processing for EHR-based pharmacovigilance: a structured review. Drug Saf. 2017;40(11):1075–89.
  44. Mamoshina P, Bueno-Orovio A, Rodriguez B. Dual transcriptomic and molecular machine learning predicts all major clinical forms of drug cardiotoxicity. Front Pharmacol. 2020;11:639.
  45. Ebrahimian S, Kalra MK, Agarwal S, et al. FDA-regulated AI algorithms: trends, strengths, and gaps of validation studies. Acad Radiol. 2022;29(4):559–66.
  46. European Commission. Proposal for a Regulation laying down harmonised rules on artificial intelligence (Artificial Intelligence Act). 2021.
  47. Liu Q, Huang R, Hsieh J, et al. Landscape analysis of the application of artificial intelligence and machine learning in regulatory submissions for drug development from 2016 to 2021. Clin Pharmacol Ther. 2023;113(4):771–4.
  48. Liu F, Panagiotakos D. Real-world data: methods, applications, challenges and opportunities. BMC Med Res Methodol. 2022;22:287.
  49. Labovitz DL, Shafner L, Reyes Gil M, Virmani D, Hanina A. Using artificial intelligence to reduce the risk of nonadherence in patients on anticoagulation therapy. Stroke. 2017;48(5):1416–9.
  50. Barbieri C, Molina M, Ponce P, et al. AI clinical decision support optimizes anemia management in hemodialysis patients. Kidney Int. 2016;90(2):422–9.
  51. Damiani G, Altamura G, Zedda M, et al. Potentiality of algorithms and AI adoption to improve medication management in primary care: a systematic review. BMJ Open. 2023;13:e065301.
  52. Paul D, Sanap G, Shenoy S, Kalyane D, Kalia K, Tekade RK. Artificial intelligence in drug discovery and development. Drug Discov Today. 2021 Jan;26(1):80–93.
  53. Fang J, Zhang P, Wang Q, et al. AI framework identifies candidate targets for drug repurposing in Alzheimer’s disease. Alzheimers Res Ther. 2022;14:7.
  54. Saednia K, Lagree A, Alera MA, et al. Quantitative digital histopathology and machine learning to predict pathological complete response to chemotherapy in breast cancer. Sci Rep. 2022;12(1):9690.
  55. Lu H, Arshad M, Thornton A, et al. A mathematical descriptor of tumor mesoscopic structure from CT images annotates prognostic and molecular phenotypes of epithelial ovarian cancer. Nat Commun. 2019;10:764.
  56. Tricco AC, Zarin W, Lillie E, et al. Utility of social media and crowd-intelligence data for pharmacovigilance: a scoping review. BMC Med Inform Decis Mak. 2018;18(1):38.
  57. Comfort S, Perera S, Hudson Z, et al. Sorting through the safety data haystack: using ML to identify ICSRs in social digital media. Drug Saf. 2018;41(6):579–90.
  58. Doudesis D, Lee KK, Boeddinghaus J, et al. Machine learning for diagnosis of myocardial infarction using cardiac troponin concentrations. Nat Med. 2023;29(5):1201–10.
  59. Guo A, Pasque M, Loh F, Mann DL, Payne PRO. Heart failure diagnosis, readmission, and mortality prediction using machine learning and AI models. Curr Epidemiol Rep. 2020;7(4):212–9.
  60. Poplin R, Varadarajan A V, Blumer K, et al. Prediction of cardiovascular risk factors from retinal fundus photographs via deep learning. Nat Biomed Eng. 2018;2(3):158–64.
  61. Mitani A, Huang A, Venugopalan S, et al. Detection of anaemia from retinal fundus images via deep learning. Nat Biomed Eng. 2020;4(1):18–27.
  62. Shamout FE, Shen Y, Wu N, et al. An artificial intelligence system for predicting the deterioration of COVID-19 patients in the emergency department. NPJ Digit Med. 2021;4:1–11.
  63. Wang Z, Ong CLJ, Fu Z. AI models to assist vancomycin dosage titration. Front Pharmacol. 2022;13:801928.
  64. Wasylewicz A, van de Burgt B, Weterings A, et al. Identifying adverse drug reactions from free-text electronic hospital health record notes. Br J Clin Pharmacol. 2022;88(3):1235–45.
  65. Rozenblum R, Rodriguez-Monguio R, Volk LA, et al. Using a machine learning system to identify and prevent medication prescribing errors: a clinical and cost analysis evaluation. Jt Comm J Qual Patient Saf. 2020;46(1):3–10.
  66. Shah AD, Bartlett JW, Carpenter J, Nicholas O, Hemingway H. Comparison of random forest and parametric imputation models for imputing missing data using MICE: a CALIBER study. Am J Epidemiol. 2014;179(6):764–74.
  67. Oakden-Rayner L, Dunnmon J, Carneiro G, Ré C. Hidden stratification causes clinically meaningful failures in ML for medical imaging. 2019.
  68. Finlayson SG, Subbaswamy A, Singh K, et al. The clinician and dataset shift in artificial intelligence. New England Journal of Medicine. 2021;385(3):283–6.
  69. Mundhenk TN, Chen BY, Friedland G. Efficient saliency maps for explainable AI. 2020.
  70. Vokinger KN, Feuerriegel S, Kesselheim AS. Mitigating bias in machine learning for medicine. Communications Medicine. 2021;1:1–3.
  71. Panch T, Mattie H, Atun R. Artificial intelligence and algorithmic bias: implications for health systems. J Glob Health. 2019;9:20318.
  72. Price WN, Cohen IG. Locating liability for medical AI. 2023.
  73. FDA CDRH. Artificial intelligence and machine learning in software as a medical device. 2022.
  74. Veinot TC, Mitchell H, Ancker JS. Good intentions are not enough: how informatics interventions can worsen inequality. Journal of the American Medical Informatics Association. 2018;25(8):1080–8.
  75. Kumar Y, Gupta S, Singla R, Hu YC. A systematic review of artificial intelligence techniques in cancer prediction and diagnosis. Archives of Computational Methods in Engineering. 2022;29(4):2043–70.
  76. Zhavoronkov A, Vanhaelen Q, Oprea TI. Will artificial intelligence for drug discovery impact clinical pharmacology? Clin Pharmacol Ther. 2020;107(4):780–5.
  77. European Parliament Research Services. Artificial Intelligence in Healthcare: Applications, Risks, and Ethical and Societal Impacts. 2022.

Photo
Dr. Snehashis Singha
Corresponding author

Resident Doctor, Department of Pharmacology and Therapeutics, King George’s Medical University, Lucknow, Uttar Pradesh, India, 226003

Photo
Dr. Rajasree Majumder
Co-author

Resident Doctor, Department of Microbiology, King George’s Medical University, Lucknow, Uttar Pradesh, India, 226003

Dr. Snehashis Singha, Dr. Rajasree Majumder, Artificial Intelligence in Pharmacology: From Drug Discovery to Personalized Medicine and Future Horizons, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 10, 648-669. https://doi.org/10.5281/zenodo.17285741

More related articles
Artificial Intelligence in Pharmacy: Current Role,...
Harsh More , Aryan Maurya , Neelam Yadav, Kajal Shirapure , Dr. V...
Artificial Intelligence in Cancer Biomedicine: A C...
Brijesh Yadav , Vanshika , Vinay Pal Chaudhary , Ashu Kumar, Mant...
From Genes to Drugs: Pharmacogenomic Strategies for Personalized Therapy in Indi...
Divya Tayade , Priyanshu Giripunje , Punam Jagnit, Nupur Falke, Lavanya Dhamne, Astha Nagrare, Nives...
The Role of Artificial Intelligence and Machine Learning in Modern Drug Discover...
Dr. Santosh Dighe, Saurabh Shinde, Siddhi Shinde, Atharv Vike, ...
Artificial Intelligence and Machine Learning in Novel Drug Delivery Systems (NDD...
Shyam Manza, Shweta Galande, Srushti Bansode, Sushma Waghmare, Sonai Antapure, Priyatama Powar, ...
Related Articles
Artificial Intelligence in the Pharmaceutical Industry: A Comprehensive Review...
Bharat Jadhav, Dr. Hemant Gangurde, Ketan Deshmukh, Ravidas Dhakane, Nilesh Dhole, ...
Artificial Intelligence in Pharmaceutical Analysis: A Review...
K. Bhavyasri, C. A. Sri Ranjani, Syeda Farhat Sultana, ...
More related articles
Artificial Intelligence in Pharmacy: Current Role, Future Potential & Transforma...
Harsh More , Aryan Maurya , Neelam Yadav, Kajal Shirapure , Dr. Vinod Bairagi, ...
Artificial Intelligence in Cancer Biomedicine: A Comprehensive Survey of Diagnos...
Brijesh Yadav , Vanshika , Vinay Pal Chaudhary , Ashu Kumar, Mantasha Rani, ...
Artificial Intelligence in Pharmacy: Current Role, Future Potential & Transforma...
Harsh More , Aryan Maurya , Neelam Yadav, Kajal Shirapure , Dr. Vinod Bairagi, ...
Artificial Intelligence in Cancer Biomedicine: A Comprehensive Survey of Diagnos...
Brijesh Yadav , Vanshika , Vinay Pal Chaudhary , Ashu Kumar, Mantasha Rani, ...