Next Article in Journal
Tanacetum balsamita L.: Botany, Traditional Uses, Phytochemical Profiling, and Biological Activities
Previous Article in Journal
Pharmacological Applications of Electrospun Nanofibers Loaded with Bioactive Natural Compounds and Extracts: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Review on Revolutionizing Healthcare Technologies with AI and ML Applications in Pharmaceutical Sciences

Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be University), Pune 411038, India
*
Author to whom correspondence should be addressed.
Drugs Drug Candidates 2025, 4(1), 9; https://doi.org/10.3390/ddc4010009
Submission received: 30 January 2025 / Revised: 21 February 2025 / Accepted: 26 February 2025 / Published: 4 March 2025

Abstract

:
Background/Objectives: The integration of Artificial Intelligence (AI) and Machine Learning (ML) in pharmaceutical research and development is transforming the industry by improving efficiency and effectiveness across drug discovery, development, and healthcare delivery. This review explores the diverse applications of AI and ML, emphasizing their role in predictive modeling, drug repurposing, lead optimization, and clinical trials. Additionally, the review highlights AI’s contributions to regulatory compliance, pharmacovigilance, and personalized medicine while addressing ethical and regulatory considerations. Methods: A comprehensive literature review was conducted to assess the impact of AI and ML in various pharmaceutical domains. Research articles, case studies, and industry reports were analyzed to examine AI-driven advancements in predictive modeling, computational chemistry, clinical trials, drug safety, and supply chain management. Results: AI and ML have demonstrated significant advancements in pharmaceutical research, including improved target identification, accelerated drug discovery through generative models, and enhanced structure-based drug design via molecular docking and QSAR modeling. In clinical trials, AI streamlines patient recruitment, predicts trial outcomes, and enables real-time monitoring. AI-driven predictive maintenance, process optimization, and inventory management have enhanced efficiency in pharmaceutical manufacturing and supply chains. Furthermore, AI has revolutionized personalized medicine by enabling precise treatment strategies through genomic data analysis, biomarker discovery, and AI-driven diagnostics. Conclusions: AI and ML are reshaping pharmaceutical research, offering innovative solutions across drug discovery, regulatory compliance, and patient care. The integration of AI enhances treatment outcomes and operational efficiencies while raising ethical and regulatory challenges that require transparent, accountable applications. Future advancements in AI will rely on collaborative efforts to ensure its responsible implementation, ultimately driving the continued transformation of the pharmaceutical sector.

1. Introduction

Artificial Intelligence (AI) refers to techniques used to build systems that emulate human intelligence. It encompasses various components of intelligence, including reasoning, learning, problem-solving, perception, and linguistic understanding [1].
Machine Learning (ML), a subset of AI, focuses on developing algorithms and statistical models. These models learn from data and make predictions or decisions without explicit instructions. During training, ML models acquire knowledge from data, drawing from probability theory and linear algebra. Key parameters and processes required to implement a machine learning model have been summarized in Table 1. There are two main types of ML:
  • Supervised Machine Learning: In supervised ML, both the data and the problem is known. When given a set of features (x), we can predict the value of y. It includes both classification (assigning data to categories) and regression (predicting numerical values).
  • Unsupervised Machine Learning: The data provided are unlabeled. Clustering groups these data points together (useful for detecting anomalies or creating new categories). Dimension reduction can help in visualize complex datasets [2].
Table 1. Key parameters and processes involved in implementing machine learning models.
Table 1. Key parameters and processes involved in implementing machine learning models.
ParametersDescriptionAI/ML Models UsedWorkingReferences
Data CollectionElectronic Health Records (EHRs), Genomic Data, Imaging Data, Clinical Data, Lifestyle Data, Environmental DataN/AThe collection of data from various sources is the first step in precision medicine. These data are used to develop personalized treatment plans for individual patients.[3,4,5,6]
Data PreprocessingData Cleaning, Data Integration, Data Transformation, Data ReductionN/AThe preprocessing of data is an essential step in precision medicine. This involves cleaning and transforming the data to make them suitable for analysis.[3,4,5,6]
Machine Learning Model SelectionSupervised Learning (Classification, Regression), Unsupervised Learning (Clustering), Reinforcement LearningRandom Forest, Support Vector Machine, Neural Networks, Decision Trees, Naive Bayes, K-Nearest Neighbors, Gradient Boosting, Deep Learning, Convolutional Neural Networks (CNN), Recurrent Neural Networks (RNN)The selection of the appropriate machine learning model is crucial in precision medicine. Supervised learning models are used for classification and regression tasks, while unsupervised learning models are used for clustering tasks. Reinforcement learning models are used for decision-making tasks.[7]
Model TrainingModel Selection, Model Training, Model Evaluation, Model OptimizationRandom Forest, Support Vector Machine, Neural Networks, Decision Trees, Naive Bayes, K-Nearest Neighbors, Gradient Boosting, Deep Learning, Convolutional Neural Networks (CNN), Recurrent Neural Networks (RNN)The training of the machine learning model is an essential step in precision medicine. This involves selecting the appropriate model, training it on the pre-processed data, and evaluating its performance. The model is then optimized to improve its accuracy.[8]
Model DeploymentModel Integration with EHRs, Model Integration with Clinical Workflows, Model Integration with Imaging SystemsRandom Forest, Support Vector Machine, Neural Networks, Decision Trees, Naive Bayes, K-Nearest Neighbors, Gradient Boosting, Deep Learning, Convolutional Neural Networks (CNN), Recurrent Neural Networks (RNN)The deployment of the machine learning model is the final step in precision medicine. This involves integrating the model with EHRs, clinical workflows, and imaging systems to make it accessible to healthcare providers.[9]
Model Monitoring and UpdatingModel Performance Monitoring, Model Updating with New Data, Model Retraining with New DataRandom Forest, Support Vector Machine, Neural Networks, Decision Trees, Naive Bayes, K-Nearest Neighbors, Gradient Boosting, Deep Learning, Convolutional Neural Networks (CNN), Recurrent Neural Networks (RNN)The monitoring and updating of the machine learning model are essential in precision medicine. This involves monitoring the model’s performance, updating it with new data, and retraining it with new data to ensure that it remains accurate and up-to-date.[10,11]
N/A: No AI models were used in this particular step of the process as part of implementing machine learning models.
Additionally, Deep Learning, a subset of ML, relies on multilayered neural networks to solve tasks. It has revolutionized fields like computer vision, natural language processing, and speech recognition [1]. Figure 1 provides an overview of the types of machine learning algorithms.
The integration of Artificial Intelligence (AI) and Machine Learning (ML) in pharmaceutical sciences has brought about a transformative impact on various facets of the field. These cutting-edge technologies have revolutionized drug discovery, development, and patient care by leveraging advanced computational algorithms to analyze extensive datasets encompassing genomics, proteomics, and chemical structures [12]. Through the prediction of drug–target interactions, identification of novel drug candidates, and optimization of molecular compounds, AI/ML methodologies enable researchers to gain insights into disease mechanisms at a molecular level, facilitating the design of more efficacious therapies [13]. Notably, recent advancements in deep learning models, such as graph neural networks, have significantly expedited drug discovery processes by accurately predicting protein–ligand binding affinities [12].
AI/ML also plays a pivotal role in enhancing various aspects of clinical trial design and execution. These technologies aid in optimizing patient recruitment by identifying suitable candidates based on genetic profiles and clinical characteristics, thereby facilitating adaptive trial designs that minimize costs and time [14]. Real-world data (RWD) from wearable devices, electronic health records, and imaging studies are harnessed to provide valuable insights, which AI algorithms analyze to personalize treatment regimens, predict adverse events, and stratify patient populations [15]. For instance, AI-powered algorithms can predict patient responses to immunotherapies by scrutinizing tumor microenvironment features.
Moreover, in the domain of pharmacovigilance and safety surveillance, AI/ML algorithms are instrumental in post-market surveillance for monitoring drug safety. By analyzing adverse event reports, electronic health records, and social media data, these tools promptly detect safety signals, thereby enhancing pharmacovigilance by identifying rare adverse events that might otherwise go unnoticed [16]. Additionally, the application of Natural Language Processing (NLP) models facilitates the extraction of valuable information from unstructured text, enabling efficient analysis of medical literature and adverse event narratives [17].
In the context of pharmaceutical manufacturing and quality control, AI/ML technologies optimize manufacturing processes to ensure consistent drug quality [18]. Predictive maintenance models are employed to prevent equipment failures, thereby minimizing production downtime. Quality control procedures benefit from AI-driven image analysis, as machine vision systems detect defects in drug formulations, packaging, and labeling, thereby ensuring compliance with regulatory standards [19].
As AI/ML models become increasingly integral to decision-making processes, ethical considerations become paramount. Ensuring transparency and interpretability of these models is crucial, prompting researchers to develop explainable AI techniques to comprehend model predictions and address biases. Collaborative efforts among pharmaceutical scientists, clinicians, and data scientists are deemed essential for the responsible adoption of AI in healthcare.

2. Drug Discovery and Development

The journey of developing new drugs involves recognizing drug targets, validating them, progressing from hits to lead compounds, refining leads, identifying preclinical molecules, evaluating them preclinically, and conducting clinical trials to bring a new drug to market [20]. The average pre-tax expenditure to bring a new prescription drug to market is around USD 2.6 billion, spanning a period of 5.9–7.2 years for non-oncological drugs and 13–15 years for oncological drugs [12]. Despite the substantial financial commitment, the success rate for novel small drugs gaining clinical approval is only 13%, with a significant risk of failure [21]. The complex and vast data from genomics, proteomics, microarray data, and clinical trials present a significant challenge in the drug discovery pipeline [22]. The advent of computer-assisted drug design technology is viewed as a promising solution to enhance this challenging landscape by streamlining the drug development process effectively [20].
Furthermore, employing computational methods that incorporate multi-objective refinement can help decrease the failure rate of preclinical lead compounds [2]. Within the scope of drug development, artificial intelligence (AI) utilizes computer software to analyze, learn from, and interpret vast pharmaceutical data, leveraging advancements in machine learning (ML) to streamline the discovery of new drug molecules in a cohesive and automated manner [22].
Presently, AI technologies, particularly Deep Learning (DL) methodologies, show great potential in drug design because of their remarkable ability to generalize and extract features [23]. Traditional machine learning methods rely on manually designed features, whereas Deep Learning techniques can autonomously learn features from input data, transforming basic attributes into complex characteristics through multi-layer feature extraction [24]. Figure 2 aptly summarizes the various applications of AI/ML in the drug discovery field.

2.1. Predictive Modeling for Target Identification

The strategy of altering a target’s activity to combat a disease is a common approach in drug discovery. Identifying new targets that can be modulated to achieve a therapeutic effect with an acceptable safety profile is often the initial step in drug development. While there is a growing emphasis on discovering novel therapeutic targets associated with diseases, the experimental validation of these targets is both time-consuming and expensive [25,26]. To streamline the selection of the most promising target candidates for future investigations, researchers have turned to AI and machine learning (ML) techniques.
Lipinski’s Rule of Five (Ro5), established in 1997 based on Phase II drug physicochemical profiles, guides the design of developable molecules by flagging potential issues like excessive hydrogen-bond donors and acceptors, high molecular weight, and elevated Log p values [20]. While Ro5 has been instrumental in designing compounds for known targets, there is a growing need for innovation to target new biological pathways. Beyond Ro5, emerging modalities like bifunctional small molecules, peptides, and oligonucleotides offer novel avenues for drug discovery [27]. Carbohydrate-based drug research is gaining traction, with over 170 approved drugs showcasing their potential across various therapeutic areas [28]. Lipids, crucial for cellular function, present a rich source of drug targets, particularly in lipid signaling pathways and proteins [29]. Druggable proteins, essential for small molecule interactions, remain a key focus in drug development, although the landscape of potential targets is still being explored. The challenge lies in efficiently delivering therapeutic agents, with traditional computational methods facing limitations in accurately predicting interactions. The wealth of complex data from various sources further complicates the drug discovery process, necessitating innovative solutions to overcome these obstacles.
Artificial Intelligence (AI) has significantly advanced big data analytics in biomedical research by offering a wide array of Machine Learning (ML) techniques for extracting valuable insights from complex datasets. In the context of drug discovery, AI-driven models complement Ro5 by refining drug-likeness assessments and predicting molecular properties such as solubility, permeability, and metabolism with greater accuracy. AI enables the identification of exceptions to Ro5, allowing researchers to explore non-traditional drug candidates such as peptides and biologics that may not conform to classic small-molecule guidelines [10]. In target identification, gene expression features are commonly utilized to unravel disease mechanisms and pinpoint genes associated with specific disorders [30]. Repositories like NCBI Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) provide extensive gene expression data for analysis [20]. Genome-wide association studies (GWAS) play a crucial role in understanding the genetic basis of complex disorders, with platforms like GWAS Central and NHGRI-EBI GWAS Catalogue containing valuable genetic data [31,32]. For instance, Ref. [33] utilized multiple functional gene networks and a kernel-based method to prioritize genes based on disease MeSH keywords, enhancing the process of prioritizing disease-related genes.
AI tools like Google’s DeepMind’s AlphaFold, trained on protein structural data, can predict protein 3D structures from amino acid sequences [34], while text mining-based tools leverage Natural Language Processing (NLP) to extract structured data from unstructured text, aiding in traditional drug discovery processes [20]. Notably, AlphaFold has been instrumental in determining the structure of previously unsolved proteins, such as those related to SARS-CoV-2, aiding in the rapid development of antiviral treatments [35]. Additionally, AI-driven platforms like Insilico Medicine’s PandaOmics have successfully identified novel drug targets for fibrosis and cancer, accelerating preclinical research [36]. Text mining-based tools leveraging Natural Language Processing (NLP), such as IBM Watson, have also been employed in pharmaceutical research to scan vast biomedical literature and extract relevant drug-target interactions, expediting hypothesis generation for new therapeutics [37]. These AI-driven approaches hold promise in accelerating target identification, drug development, and personalized medicine in the pharmaceutical industry.

2.2. Drug Repurposing Using AI and ML Techniques

Drug repositioning in drug design and discovery involves exploring drugs that were initially developed for one disease and repurposing them for other conditions [38]. The success of repositioning drugs is attributed to their ability to interact with multiple targets across various diseases, potentially leading to enhanced clinical efficacy. By leveraging this polypharmacology phenomenon, drug repositioning aims to identify new therapeutic opportunities by reevaluating existing compounds for different medical indications [39]. This approach offers a faster and more cost-effective alternative to traditional drug development, bypassing certain phases and reducing the overall time and investment required for bringing a new drug to market [40].
The biggest challenge in drug repositioning lies in customizing and optimizing methods to develop efficient and affordable drug repositioning pipelines for complex diseases [41]. Screening methods for drug reuse are crucial, and approaches in drug repositioning can be categorized as drug-oriented, target-oriented, or disease/therapy-oriented based on available information quality and quantity. Additionally, classification can be based on network-based, ligand-based, chemogenomic, and machine learning (ML) approaches. With the rise of high-throughput technologies, computational analysis and mining tools are essential to explore vast amounts of data. In silico drug repositioning, driven by hypothesis, translates omics data into predictions of druggable targets, leveraging various data sources. AI and ML methods, utilizing publicly available databases, have significantly impacted drug discovery. For instance, DSP-1181, the first repurposed drug discovered through AI, entered clinical trials in less than 12 months, showcasing the accelerated drug development process. AI-based approaches have revolutionized drug development by rapidly identifying bioactive compounds from large candidate pools, facilitating precision medicine. Moreover, AI has enabled the creation of reverse vaccinology virtual frameworks and ML models that learn patterns from data, including deep learning for enhanced learning processes in drug discovery.
ML algorithms are replacing traditional methods like chemical similarity and molecular docking with new systems biology approaches to assess drug effects. This shift has led to the development of various AI-based algorithms and web tools such as DrugNet, DRIMC, DPDR-CPI, PHARMGKB, PROMISCUOUS 2.0, and DRRS in recent times.

2.3. Lead Optimization Through Machine Learning Algorithms

Integrating computational modeling with domain-aware machine learning (ML) models, followed by iterative experimental validation, can speed up the identification of possible therapeutic candidates. Although thousands of new candidates can be produced by generative deep learning models, their physiochemical and biochemical attributes are usually not fully optimized [42]. There are several ways ML can be applied to accelerate lead optimization:
  • Structural Alert and Toxicity Analysis: ML can be used to predict the toxicity of a compound based on its structure [43].
  • High-Throughput Virtual Screening: ML algorithms can quickly screen large databases of compounds to identify potential leads [44].
  • 3D Quantitative Structure–Activity Relationships (QSAR): ML can be used to predict the biological activity of a compound based on its 3D structure [44].
  • Multi-Parameter Optimization: ML can optimize multiple parameters simultaneously to find the best lead compounds [42].
  • Graph Neural Networks: These can be used to predict the properties of a compound based on its molecular graph [10].

2.4. De Novo Drug Design with Generative Models

A promising area in computational chemistry and drug discovery is de novo drug design using generative models. This is the process of creating new chemical compounds that may one day be used as medications using machine learning models [45].
The application of generative models for conditional graphs is one strategy. These models produce molecules by generating a series of graphs, each of which shows a potential intermediate state during the molecule’s synthesis [45]. This approach is quite flexible and works well for generation with several goals in mind. It performs better than models that produce SMILES strings, particularly in terms of the output rate that is valid [45].
A different strategy is to employ 3D deep generative models. For instance, the DeepLigBuilder programme uses an innovative graph generative model called Ligand Neural Network (L-Net) to construct 3D molecules with high drug-likeness that are both chemically and conformationally valid from start to finish. This model is used for structure-based de novo drug design tasks, in conjunction with Monte Carlo tree search [46,47].
For de novo molecular design, reinforcement learning can also be used with deep generative models. By using this method, the generative model can be adjusted to produce molecules with different sets of desirable properties [48].
These techniques have been used to address a number of drug design issues, such as the synthesis of compounds with a specified scaffold, molecules that meet particular requirements for synthetic accessibility and drug-likeness, and dual inhibitors against particular targets [26]. However, it’s crucial to remember that, despite their potential, these techniques are still in the research and development stages, and it is yet unclear how useful they will be in the actual process of finding drugs. Figure 3 demonstrates the integration of AI/ML algorithms across the drug discovery continuum.

3. Cheminformatics and Computational Chemistry

Cheminformatics, or chemoinformatics, stands at the intersection of physical chemistry theory and computer science techniques, offering a multidisciplinary approach to solving descriptive and prescriptive challenges within chemistry and its applications to related fields like biology [49]. This field is founded on the principles of effective data representation and manipulation, harnessing the power of computers to store and process chemical formulas, properties, and pertinent information. Its primary objective is to develop tools and methodologies for managing vast sets of chemical data, enabling tasks such as data mining, machine learning, and predictive modeling [50]. In parallel, computational chemistry, with its emphasis on computational methods, provides a complementary avenue for understanding molecular systems. Utilizing techniques like quantum mechanics, molecular dynamics, and density functional theory (DFT), computational chemists delve into the electronic structures, molecular dynamics, and properties of chemical compounds [51].

3.1. Virtual Screening of Compound Libraries

Discovering new scaffolds and chemotypes through high-throughput screening is incredibly challenging due to its labor-intensive and resource-heavy nature. The vast number of commercially available small molecules makes conducting comprehensive in vitro tests practically intractable [52]. Drug discovery, particularly within biopharmaceutical companies, is among the most costly, time-consuming, and complex endeavors. It involves identifying and optimizing lead compounds from large chemical libraries, which must exhibit high-affinity binding and specificity for disease-associated targets, along with favorable pharmacodynamic and pharmacokinetic properties (ADMET properties) [44]. This process represents a multivariable optimization task typically conducted on supercomputers using reliable scoring functions to assess the binding affinity or inhibition potential of potential drug-like compounds [53]. The main challenge stems from the sheer number of compounds in chemical spaces, making computational drug discovery demanding, albeit more cost-effective and time-efficient than experimental high-throughput screening. The primary objective is to identify the most stable (global) minima among numerous protein–ligand complexes, which can range from 106 to 1012 [54,55]. To tackle this challenge, the parallel implementation of in silico virtual screening is essential to ensure drug discovery within feasible timeframes. In the field of drug discovery, virtual screening of compound libraries using AI/ML techniques is rapidly evolving. Several notable approaches have been developed to streamline this process including Virtual Synthon Hierarchical Enumeration Screening (V-SYNTHES), which significantly reduces the number of molecules requiring assessment in large chemical libraries. For example, from an 11 billion-molecule library, approximately 2 million are prioritized for screening based on their 3D structure and the target site [56].
VSFlow, an open-source ligand-based virtual screening tool, incorporates substructure-, fingerprint-, and shape-based virtual screening, offering high customizability [57].
AI Accelerated Virtual Screening Platform integrates AI algorithms to efficiently screen multi-billion compound libraries against diverse targets [58].
Machine Learning on DNA-encoded libraries, demonstrated by Google Research and X-Chem, combines physical screening with DNA-encoded small-molecule libraries and virtual screening using a graph convolutional neural network (GCNN) [59].
Lig3DLens is an end-to-end computational toolbox for 3D virtual screening based on shape and electrostatics similarity to a reference (hit) compound [58]. These methodologies harness the capabilities of AI/ML to expedite the drug discovery process by effectively navigating through extensive compound libraries. They signify significant progress in the field of computational chemistry and drug design, promising enhanced efficiency and efficacy in identifying potential therapeutics.

3.2. QSAR Modeling for Predicting Compound Properties

Quantitative Structure-Activity Relationship (QSAR) modeling is a computational technique used to establish a mathematical relationship between the chemical structure of molecules and their biological or chemical activities. The fundamental idea behind QSAR is that the biological activity of a compound is closely related to its molecular structure, and by quantifying molecular features, it is possible to predict the activity of new compounds [60]. This approach is widely utilized in drug discovery, toxicology, and environmental chemistry to identify potential lead compounds, optimize existing molecules, and assess the risks of chemical substances.
QSAR models rely on the generation of molecular descriptors, which are numerical representations of a compound’s physicochemical properties, such as hydrophobicity, size, charge distribution, and shape [61]. These descriptors serve as the input variables for statistical or machine learning models, which attempt to correlate these molecular characteristics with the observed biological activity, such as potency, toxicity, or binding affinity. Common methods employed to build QSAR models include linear regression, partial least squares, support vector machines, and neural networks [61].
A key advantage of QSAR modeling is its ability to predict the activity of untested compounds, saving time and resources in experimental studies. However, the accuracy of QSAR models depends heavily on the quality and diversity of the data used to train them [62]. Proper dataset selection is essential to ensure that the models are both robust and generalizable. Furthermore, QSAR models are limited by the quality of the descriptors chosen, as they must accurately capture the molecular features that influence biological activity. Despite these challenges, QSAR remains a powerful tool in computational chemistry, enabling more efficient drug design and risk assessment processes [62].
To address challenges and enhance predictive accuracy, ensemble-based machine learning approaches have emerged as valuable solutions. These methodologies construct a diverse array of models and amalgamate their predictions, aiming to overcome limitations and bolster reliability. However, prevalent approaches like random forests and other ensemble techniques in QSAR prediction often restrict their model diversity to a single subject, prompting the need for advancements in this domain [62].
In recent developments, Auto-ML tools have garnered attention for their efficacy in molecule property prediction tasks. Notable among these is Uni-QSAR, a tool that integrates molecular representation learning (MRL) encompassing 1D sequential tokens, 2D topology graphs, and 3D conformers. By incorporating pretraining models and harnessing rich representations from extensive unlabeled data, Uni-QSAR demonstrates promising capabilities in enhancing prediction accuracy [63]. Similarly, tools such as DeepAutoQSAR and ChemProp have surfaced, offering versatile and widely utilized options in real-world applications [63].
Furthermore, the field has witnessed strides in Conformal Prediction (CP) algorithms tailored to advanced machine learning models such as Deep Neural Networks and Gradient Boosting Machines. CP represents a promising avenue due to its algorithm-agnostic nature, enabling the generation of valid prediction intervals under certain mild assumptions regarding the data distribution. Techniques such as Random Forest, Neural Networks, deep learning, and the Monte Carlo method have demonstrated success in generating robust QSAR models [61,64].
A notable strategy in QSAR modeling is the consensus approach, which aims to refine and enhance model predictions by leveraging new, advanced machine learning algorithms. This approach focuses on standardizing, streamlining, and automating various steps within the QSAR modeling process, culminating in a final prediction that synthesizes the outputs of multiple models [65].
Activity-cliff prediction is a concept addressing pairs of compounds that are structurally similar yet exhibit significant differences in binding affinity for a given target. These pairs, known as activity cliffs (ACs), pose challenges for QSAR models, often resulting in prediction errors. While it has been hypothesized that modern QSAR methods struggle with AC prediction, the quantitative relationship between AC prediction power and general QSAR performance remains an area of ongoing exploration [66].
Ensemble methods have emerged as valuable tools extensively employed in drug research within the QSAR domain. These methods encompass a diverse range of approaches, including data sampling ensembles, such as neural network ensembles based on bootstrap sampling in QSAR; method ensembles, involving ensembles against different learning methods for drug–drug interaction; qualitative and quantitative SAR models using ensemble learning; hybrid QSAR prediction models employing various learning methods; ensembles utilizing different boosting methods; hybrid approaches that combine feature selection and feature learning in QSAR modeling; and representation ensembles, leveraging ensembles against diverse chemicals for carcinogenicity prediction [62].
These ensemble methods represent a versatile and powerful collection within QSAR modeling, offering strategies to improve prediction accuracy and address complex challenges in understanding chemical–biological relationships. Figure 4 outlines the use of AI/ML during various stages of QSAR workflow.
For example, Ambure et al. have created the user-friendly standalone program “QSAR-Co” version 1.0.0, which may be downloaded for free from https://sites.google.com/view/qsar-co (accessed on 4 January 2025) [64]. The goal of this software is to create reliable QSAR models based on categorization for a wide range of data sets, even if the response data set values relate to several theoretical or experimental settings or to several biological targets. Using two well-known techniques—LDA and RF—the program offers all the necessary functionality to create classification-based QSAR models. In addition, it provides the necessary methods (such as ROC analysis and the Y-randomization test) to evaluate the robustness of the created models. These tools allow users to calculate the models’ application domain and the prediction reliability for query chemicals. All the procedures needed to construct classification-based QSAR models are carried out by only clicking a button once all the parameters and approaches have been established [64].

3.3. Molecular Docking and Dynamics Simulations

Molecular Docking involves using computational methods to forecast how one molecule will position itself in relation to another when they bind together to create a stable complex. This technique is frequently employed in the strategic development of medications, offering insights into how small molecule drug candidates bind to their protein targets [67,68].
Molecular Dynamics Simulations (MDS) offer valuable insights into the functional mechanisms of proteins, peptides, and other biomolecules. MDS can complement traditional experiments by providing a detailed understanding of biological processes at the molecular level [69].
Artificial Intelligence and Machine Learning (AI/ML) can be integrated with these techniques to improve their predictive accuracy and efficiency. For example, AI/ML can predict the binding strength between a ligand and a receptor based on features derived from docking results, expediting drug discovery by narrowing down potential compounds for lab testing [70].
In a recent investigation, scientists utilized molecular docking and dynamics simulations to explore the potential inhibitory effects of ACE inhibitors against SARS-CoV-2 targeting the hACE2 receptor. They discovered that Alacepril and Lisinopril interacted with the human angiotensin-converting enzyme 2 (hACE2), which serves as the entry point for the SARS-CoV-2 spike protein [71].
Another study employed an unsupervised deep-learning framework to analyze the more flexible SARS-CoV-2 main protease (Mpro). They conducted MD simulations of Mpro with various ligands, focusing on binding-site residues and stable protein conformations over time. The chosen optimal descriptor was the distance between residues and the center of the binding pocket. Using this method, they generated a local dynamic ensemble and utilized a neural network to compute Wasserstein distances across different system pairs, revealing ligand-induced conformational differences in Mpro [72].
Despite their promising results, it’s crucial to acknowledge that these are computational predictions that require validation through experimental data to ensure accuracy.

3.4. Structure-Based Drug Design Aided by AI

Structure-based drug design empowered by Artificial Intelligence (AI) has emerged as a rapidly advancing field in recent years, fundamentally changing the way we identify or create effective molecular structures against disease targets with desired drug properties. This integration of AI has the potential to make the drug discovery process much simpler, faster, and more cost-effective [73].
An example of this game-changing approach is the Artificial Intelligence-driven Drug Design (AIDD) platform. This innovative platform uses advanced computer algorithms to automate the process of designing new drugs. It combines simulations and predictions for how drugs are absorbed, distributed, metabolized, excreted, and their potential toxicity (known as ADMET properties) with a powerful evolutionary algorithm [74].
Instead of relying solely on traditional methods that focus on making incremental changes to existing molecular structures in order to enhance their binding to target proteins [75], the AIDD platform takes a completely different approach. It uses an iterative optimization process that takes into account multiple factors such as target affinity, activity, as well as important pharmacokinetic and ADMET properties. This allows it to generate entirely new molecules that have similar properties to known lead compounds [76].

4. Clinical Trials Optimization

4.1. Patient Recruitment and Eligibility Assessment Using AI Algorithms

AI has the potential to greatly improve patient recruitment and eligibility assessment in clinical trials. By analyzing large datasets, AI can identify specific groups of patients who may benefit more from certain treatments. This helps researchers target their recruitment efforts towards those who are most likely to respond positively to the intervention [77].
AI algorithms can extract useful information from social media posts, such as discussions about symptoms or experiences with a particular condition. These data can then be used to identify geographical areas where the condition is more prevalent, making it easier to find suitable participants for a clinical trial. Instead of manually reviewing hundreds or even thousands of electronic health records, AI can quickly scan through them to identify individuals who meet the eligibility criteria for a study [78]. This saves time and resources for both researchers and healthcare providers. Once potential participants have been identified, AI systems can automatically send notifications to both the healthcare provider and the patient, informing them about the opportunity to join a clinical trial [78]. This proactive approach helps ensure that eligible individuals are aware of the option and can consider participating. Figure 5 shows various AI tools and their uses across the clinical trial process.
In some cases, the eligibility criteria for a clinical trial can be quite complex and difficult for patients to understand. AI technologies can help simplify these criteria and present them in a more user-friendly manner, making it easier for individuals to determine if they qualify for a study. AI-powered solutions like chatbots or virtual assistants can interact with potential trial participants, providing them with information about the study, answering their questions, and even collecting initial screening data [79]. This improves patient engagement and increases the likelihood of individuals following through with the enrolment process.
NLP is a branch of AI that enables computers to understand and interpret human language. In the context of clinical trials, NLP can be used to analyze doctors’ notes or pathology reports, automatically identifying patients who meet the criteria for a specific study.

4.2. Predictive Analytics for Trial Outcome Prediction

The results of clinical trials can be predicted using AI analytics. For example, an algorithm known as HINT (hierarchical interaction network) developed by computer scientist Jimeng Sun’s team at the University of Illinois Urbana-Champaign can predict the success of a clinical trial based on the drug molecule, target disease, and patient eligibility. They then developed a technique known as SPOT (sequential prediction modeling of clinical trial outcome), which takes into account more recent studies and also considers the dates of the trials in its training data [80].

4.3. Real-Time Monitoring of Patient Data for Safety and Efficacy Analysis

Real time monitoring of the patient data is important to ensure safety and data integrity during clinical trials. AI can be used for Risk-based management (RBM) for the same. RBM focusses on monitoring the trial processes which are most likely to affect the safety of patients and legitimacy of data, by using real time analysis [12]. AI algorithms can also monitor adverse events, anomalies and laboratory results and effectively report them.

4.4. Personalized Medicine and Treatment Response Prediction

The ability of AI algorithms to analyze large scale genomic data, treatment response and clinical outcomes can be utilized further to predict individual treatment response and progress [81]. This can enable the healthcare providers to select optimal therapies and minimize the risk of adverse events. AI can also analyze the lifestyle factors, unique genetic makeup and identify the subgroups of a populations which are most likely to respond to a particular treatment [82].

5. Fundamentals of Perturbation-Theory Machine Learning (PTML)

Perturbation-Theory Machine Learning (PTML) is an advanced computational approach that enhances traditional machine learning (ML) techniques by incorporating perturbation theory principles. It is designed to overcome the limitations of conventional AI models in pharmaceutical sciences by enabling multi-target predictions, improving model interpretability, and optimizing multiple endpoints simultaneously [83].
PTML works by integrating perturbation theory—a mathematical framework that evaluates small changes in a system—to machine learning models. The key principles include:
  • Multi-Target Learning: Unlike conventional AI models that focus on a single target (e.g., a protein or a specific disease pathway), PTML simultaneously predicts interactions across multiple biological targets, making it more suitable for complex, multi-genetic diseases [84].
  • Physicochemical and Structural Interpretability: PTML allows for a deeper understanding of molecular features that contribute to biological activity, reducing the black-box nature of AI models [83].
  • Multi-Objective Optimization: Most pharmaceutical applications involve optimizing multiple properties (e.g., efficacy, toxicity, and pharmacokinetics). PTML achieves this by considering multiple endpoints simultaneously [85].
Table 2 summarizes key PTML applications across anticancer, antimicrobial, and dual-target drug discovery:

6. Regulatory Compliance and Drug Safety

6.1. AI Applications in Pharmacovigilance for Adverse Event Detection

Artificial intelligence, specifically machine learning, is now more commonly utilized in pharmacovigilance to identify adverse drug events (ADEs) and adverse drug reactions (ADRs). AI algorithms are able to analyze large amounts of data from electronic health records, research articles, and social media to detect potential adverse events with greater speed and accuracy compared to traditional methods [100]. The primary applications of AI in patient safety and pharmacovigilance include identifying ADEs and ADRs, analyzing safety reports and clinical narratives, and predicting the impacts of drug–drug interactions.

6.2. Automated Compliance Monitoring and Reporting

Ensuring compliance and reporting in pharmacovigilance is essential. Real-time monitoring tools automate data collection and reporting, guaranteeing precise and uniform data entries. Additionally, these systems can prioritize safety signals according to severity and potential impact, simplifying the risk assessment and management process. Compliance monitoring in clinical trials involves proper reporting of adverse events, adherence to protocols, and informed consent procedures. In post-marketing pharmacovigilance, it includes monitoring adverse event reports from healthcare professionals and consumers, literature screening, and signal detection [101].

6.3. Risk Assessment and Mitigation Strategies Using ML Techniques

Machine learning techniques are currently being utilized to dynamically recognize, evaluate, and address risks associated with the use of AI and ML features. A risk-focused strategy for AI and ML has been suggested, drawing on recent advancements in AI methods and principles to identify and evaluate the complete risk profile for a particular use case over its lifespan. This methodology enables ongoing management of the intricacies of ML-related risks throughout the entire system lifespan, from conception to implementation [101,102].

6.4. Enhancing Drug Safety Profiles Through AI-Driven Approaches

Artificial intelligence (AI) is revolutionizing pharmacovigilance and ensuring the safety of drugs and medical devices through streamlined and enhanced analysis of safety data. AI enables the early identification of adverse events and more accurate risk assessment, ultimately improving patient outcomes. By leveraging various data sources, AI allows for proactive monitoring and mitigation of safety issues, leading to the enhancement of drug candidate safety and efficacy profiles. Real-time monitoring of trial participants and personalized adjustments to dosage and treatment regimens based on individual patient responses are facilitated by the use of AI technology [103,104].

7. Manufacturing and Supply Chain Management

7.1. Predictive Maintenance of Manufacturing Equipment

Predictive maintenance revolutionizes manufacturing operations by harnessing the power of machine learning algorithms to anticipate equipment failures before they occur. By leveraging data from sensors embedded within manufacturing equipment, predictive maintenance systems analyze patterns and anomalies to forecast potential malfunctions. The proactive scheduling of maintenance activities based on predictive insights minimizes downtime, enhances operational efficiency, and reduces maintenance costs. Moreover, predictive maintenance fosters a culture of preventive care, prolonging equipment lifespan and ensuring uninterrupted production cycles [105].

7.2. Optimization of Production Processes with Machine Learning

Machine learning drives optimization in production processes by extracting actionable insights from complex datasets and facilitating data-driven decision-making. By integrating data from diverse sources, including sensors, machines, and production systems, machine learning models uncover hidden patterns and correlations to identify opportunities for process improvement. These models leverage historical data to predict optimal process parameters and recommend adjustments in real-time to enhance efficiency and quality. Through continuous monitoring and iterative optimization, machine learning empowers manufacturers to streamline operations, minimize waste, and maximize throughput, ultimately driving competitive advantage in dynamic market environments [106,107].

7.3. Demand Forecasting and Inventory Management Using AI

AI-powered demand forecasting and inventory management systems empower organizations to navigate volatile market dynamics and optimize supply chain operations. By analyzing vast datasets encompassing sales, customer behavior, and market trends, AI models forecast future demand with unprecedented accuracy. This predictive foresight enables proactive inventory management, ensuring optimal stock levels to meet customer demand while minimizing excess inventory and associated costs. Moreover, AI-driven demand forecasting enhances supply chain resilience by facilitating agile response to shifting market dynamics and mitigating risks of stockouts or overstocking. By leveraging advanced algorithms and real-time data analytics, organizations gain a competitive edge in anticipating market demand and optimizing inventory management strategies [108].

7.4. Supply Chain Optimization for Timely Delivery of Pharmaceutical Products

Supply chain optimization emerges as a cornerstone of efficient pharmaceutical operations, facilitating timely delivery of critical healthcare products while minimizing costs and mitigating risks. By harnessing AI technologies, pharmaceutical companies optimize supply chain operations across the entire value chain, from raw material sourcing to distribution logistics. AI-driven supply chain optimization leverages predictive analytics and prescriptive modeling to optimize inventory levels, streamline transportation routes, and mitigate supply chain disruptions. Through real-time monitoring and adaptive decision-making, AI-enabled supply chain systems enhance responsiveness to dynamic market demands and ensure timely delivery of pharmaceutical products to healthcare providers and patients. Figure 6 summarizes various applications of blockchain in the pharmaceutical industry. By embracing AI-driven supply chain optimization, pharmaceutical companies navigate complexities inherent in global supply chains, enhance operational resilience, and deliver superior healthcare outcomes to patients worldwide [109].

8. Precision Medicine and Healthcare

Precision medicine, a paradigm shift in healthcare, emphasizes tailoring medical treatments to the individual characteristics of each patient. This approach leverages advancements in genomic sequencing, biomarker discovery, and artificial intelligence (AI) to revolutionize disease diagnosis, prognosis, and treatment. In this section, we delve into the various facets of precision medicine and how AI-driven technologies are reshaping healthcare delivery [81]. Specific parameters relevant to precision medicine along with the ML algorithms employed for them are summarized in Table 3.

8.1. Genomic Data Analysis for Personalized Treatment Strategies

Genomic data analysis stands at the forefront of precision medicine, offering insights into the genetic underpinnings of diseases and enabling personalized treatment strategies. The process entails the collection of genomic data from patients, which is then subjected to rigorous preprocessing to eliminate noise and artifacts. Subsequently, machine learning algorithms, trained on annotated datasets, unravel intricate genetic markers associated with specific diseases or treatment responses [3].
Through supervised learning techniques, these algorithms discern patterns within genomic data, correlating genetic variations with disease susceptibility, prognosis, and therapeutic outcomes. By identifying actionable genetic insights, clinicians can tailor treatment regimens to match the unique genetic profiles of individual patients, optimizing efficacy and minimizing adverse effects. Furthermore, the deployment of these algorithms facilitates real-time analysis of new genomic data, empowering clinicians with timely and informed decision-making capabilities [110].

8.2. AI-Driven Diagnostics and Biomarker Discovery

AI-driven diagnostic tools represent a cornerstone of precision medicine, enabling the rapid and accurate identification of disease biomarkers from diverse medical datasets. By harnessing machine learning algorithms, these tools analyze multifaceted data sources, including medical images and clinical records, to uncover subtle biomolecular signatures indicative of disease states. The process entails meticulous preprocessing of raw data to enhance signal-to-noise ratios and mitigate confounding factors [111].
Supervised learning methodologies empower these algorithms to discern nuanced patterns within complex datasets, facilitating disease diagnosis with unprecedented accuracy and efficiency. Through iterative refinement and validation, AI-driven diagnostic models augment clinicians’ diagnostic acumen, enabling earlier disease detection and intervention. Moreover, the integration of these models into clinical workflows fosters seamless data-driven decision-making, enhancing patient care and clinical outcomes [4,112].

8.3. Drug Response Prediction Based on Patient Genetics and Biomarkers

Predicting drug responses based on patient genetics and biomarkers heralds a transformative approach to pharmaceutical development and personalized medicine. Machine learning algorithms, trained on comprehensive patient datasets encompassing genomic and clinical variables, elucidate intricate relationships between genetic predispositions, biomolecular markers, and drug efficacy. Robust preprocessing techniques harmonize disparate data modalities, ensuring data integrity and minimizing spurious correlations.
Supervised learning paradigms empower these algorithms to extrapolate predictive models capable of forecasting individualized drug responses with remarkable precision. Ref. [111] By harnessing these predictive insights, clinicians can tailor treatment regimens to match patients’ genetic susceptibilities and biomarker profiles, maximizing therapeutic efficacy while minimizing adverse reactions. Furthermore, these models expedite clinical trial design and optimization, accelerating drug development timelines and ushering in a new era of precision pharmacotherapy [3].

8.4. Integration of AI and ML in Patient Care for Better Treatment Outcomes

The integration of AI and ML technologies into patient care holds immense promise for enhancing treatment outcomes and transforming healthcare delivery. By analyzing vast repositories of patient data, encompassing genomic profiles, clinical histories, and imaging studies, AI-driven models unearth latent patterns and correlations imperceptible to human observers. These insights enable risk stratification, early disease detection, and the formulation of tailored treatment plans tailored to individual patient needs [113].
For instance, ML models can dissect genomic data to discern genetic variants predisposing individuals to certain diseases, facilitating proactive interventions and personalized risk management strategies. Moreover, AI algorithms adeptly analyze medical images to identify subtle pathological features indicative of disease progression or treatment response, empowering clinicians with actionable diagnostic insights. By seamlessly integrating these AI-driven tools into electronic health records (EHRs) and clinical workflows, healthcare providers can make data-informed decisions, optimize resource allocation, and ultimately improve patient outcomes [4,113]. Table 4 provides an overview of the current landscape of various ML algorithms employed by companies in precision medicine, whereas the respective applications of these algorithms are shown in Table 5.

9. Ethical and Regulatory Considerations

As artificial intelligence (AI) and machine learning (ML) continue to permeate pharmaceutical research and healthcare, it is imperative to address the ethical and regulatory considerations inherent in their deployment. From safeguarding data privacy to ensuring algorithmic transparency and fairness, navigating the ethical landscape of AI-driven innovations requires a concerted effort to uphold ethical principles and regulatory standards [114].

9.1. Ethical Implications of AI and ML in Pharmaceutical Research

The integration of AI and ML in pharmaceutical research raises multifaceted ethical concerns, encompassing data privacy, bias mitigation, and algorithmic transparency. AI-based diagnostic tools, reliant on vast datasets for training and validation, confront ethical dilemmas regarding the handling of sensitive patient information and the potential for algorithmic bias. To mitigate these concerns, researchers and developers must prioritize ethical data collection practices, incorporating robust consent mechanisms and anonymization techniques to protect patient privacy. Moreover, ensuring algorithmic transparency and interpretability fosters trust and accountability, facilitating the identification and rectification of biased predictions [115,116]. A study by Pedraza et al. [117] discusses the ethical considerations and legislative frameworks for integrating AI and big data in pharmaceutical development. The use of AI in pharmaceuticals and biomedical sectors faces challenges such as the need for vast, reliable data, and the “Black Box” phenomenon, which makes the decision-making processes of AI models difficult to decipher. Ethical issues, particularly those impacting patient health, are also critical. The FDA is developing a risk-based strategic framework for incorporating AI within the medical products sector, emphasizing safety, efficacy, and regulatory adherence. The European Parliament’s Artificial Intelligence Act, ratified in March 2024, aims to ensure safe AI use while promoting innovation and protecting fundamental rights. This Act impacts pharmaceutical firms, especially those developing AI-driven medical devices, by requiring adherence to regulatory standards based on risk classification. It emphasizes the importance of high-quality data, algorithm validation, and continuous learning. The FDA’s Good Machine Learning Practice (GMLP) guidelines and EMA’s evolving policies on AI in medicine provide a structured approach for compliance [118]. The legislation fosters a trustworthy AI ecosystem, ensuring patient safety, data privacy, and system reliability, while motivating the pharmaceutical industry to engage in ethical AI research and development, aiming for better health outcomes and innovation [117].

9.2. Regulatory Challenges and Guidelines for AI-Driven Drug Development

AI-driven drug development presents regulatory challenges pertaining to safety, efficacy, and data privacy, necessitating stringent adherence to regulatory guidelines. Regulatory bodies, such as the FDA, have issued guidelines mandating the rigorous assessment of AI-based systems to ensure patient safety and data integrity. Compliance with regulatory requirements demands comprehensive validation studies, robust documentation, and transparent reporting of AI-driven methodologies. Moreover, safeguarding data privacy throughout the drug development lifecycle entails the implementation of encryption protocols, data anonymization strategies, and stringent access controls to mitigate privacy risks and comply with data protection regulations [119,120].

9.3. Ensuring Transparency, Fairness, and Accountability in AI Algorithms

Transparency, fairness, and accountability are pivotal considerations in the deployment of AI algorithms, particularly in healthcare settings where algorithmic decisions impact patient care outcomes. Machine learning algorithms utilized for drug response prediction, for instance, must exhibit transparency and explainability to engender clinician trust and facilitate informed decision-making. Addressing algorithmic bias requires diligent testing and validation to identify and rectify discriminatory patterns. Moreover, establishing clear lines of responsibility and oversight ensures algorithmic accountability, fostering a culture of ethical AI deployment and risk mitigation [121,122]. Model interpretability is another crucial factor because the “Black Box” nature of many AI models makes it difficult for stakeholders to understand and trust their decision-making processes [123]. This lack of transparency can hinder the adoption of AI technologies in clinical settings, as healthcare professionals need to comprehend how AI reaches its conclusions to integrate it effectively into their workflows [124]. Explainable AI (XAI) techniques like SHAP (Shapley Additive Explanations) and LIME (Local Interpretable Model-Agnostic Explanations) have been introduced to improve interpretability and facilitate regulatory approval [125].
On the other hand, data quality is equally critical, as AI systems require vast amounts of high-quality, reliable data to function accurately. Poor data quality can lead to biased or incorrect predictions, which can have serious implications for patient safety and treatment outcomes [126]. Ensuring data integrity involves stringent standards for data collection, storage, and processing, which are essential for the successful implementation of AI in pharmaceuticals.

9.4. Addressing Data Privacy Concerns in Healthcare AI Applications

Data privacy concerns loom large in healthcare AI applications, necessitating robust measures to safeguard patient confidentiality and comply with data protection regulations. Encryption techniques, anonymization protocols, and stringent access controls fortify data security, mitigating the risk of unauthorized access or data breaches. Furthermore, obtaining informed consent from patients ensures transparency and empowers individuals to make informed decisions regarding their data usage. By prioritizing data privacy and security, healthcare organizations can instill trust and confidence in AI-driven technologies while upholding ethical principles and regulatory mandates [19,127].

10. Future Perspectives and Challenges

As the pharmaceutical industry continues to evolve, the integration of artificial intelligence (AI) and machine learning (ML) holds immense promise for innovation and advancement. However, alongside these opportunities, significant challenges must be addressed to realize the full potential of AI in pharmaceutical research and healthcare [70].

10.1. Emerging Trends in AI and ML for Pharmaceutical Innovation

In recent years, AI and ML have emerged as pivotal tools in pharmaceutical innovation, catalyzing breakthroughs in drug discovery and clinical trial optimization. Deep learning algorithms, in particular, have garnered attention for their ability to decipher complex biological data and expedite drug candidate identification. Notably, companies like In-silico Medicine have leveraged deep learning models to expedite drug discovery pipelines, yielding promising results across diverse therapeutic areas. Moving forward, the convergence of AI and ML is expected to drive further innovations, with advancements in predictive modeling, virtual screening, and target identification poised to revolutionize pharmaceutical research paradigms [5,128].

10.2. Potential Impact of AI on the Future of Drug Discovery and Healthcare

The transformative potential of AI in drug discovery and healthcare is profound, offering unprecedented opportunities to expedite development timelines, reduce costs, and enhance patient outcomes. AI-based diagnostic tools, empowered by deep learning architectures, hold the promise of revolutionizing disease diagnosis by streamlining workflows, accelerating time-to-diagnosis, and improving diagnostic accuracy. Moreover, AI-driven drug development systems have the potential to reshape clinical trial paradigms by optimizing patient recruitment, stratification, and monitoring, thereby expediting the translation of promising therapeutics from bench to bedside. As AI continues to mature, its integration into healthcare ecosystems is poised to redefine standards of care, ushering in an era of personalized medicine and data-driven clinical decision-making [116,129,130].

10.3. Addressing Challenges Such as Data Quality, Interpretability, and Scalability

While AI and ML offer unprecedented opportunities, they also pose challenges related to data quality, interpretability, and scalability. The reliance on large, heterogeneous datasets raises concerns regarding data quality, as noisy or incomplete data can compromise the accuracy and reliability of AI-driven analyses. Moreover, the opacity of complex ML models can hinder interpretability, impeding clinicians’ ability to understand and trust algorithmic outputs. To mitigate these challenges, pharmaceutical companies must prioritize data cleaning and validation protocols while fostering the development of interpretable ML frameworks. Additionally, efforts to enhance scalability through the optimization of algorithms and infrastructure are imperative to accommodate the burgeoning demand for AI-driven solutions in pharmaceutical research and healthcare delivery [131].

10.4. Collaborative Efforts to Advance AI Technology in the Pharmaceutical Sector

Collaboration between industry stakeholders, academia, and regulatory bodies is essential to advancing AI technology in the pharmaceutical sector. Initiatives such as the AI Drug Discovery Consortium exemplify the power of collaborative efforts in driving innovation and overcoming shared challenges. By fostering knowledge exchange, data sharing, and cross-disciplinary partnerships, collaborative endeavors accelerate the development and deployment of AI solutions while ensuring adherence to rigorous standards of data integrity, interpretability, and scalability. Moreover, collaborative frameworks facilitate the cultivation of diverse perspectives and expertise, enriching the collective understanding of AI’s potential and its application in addressing complex healthcare challenges [76,115].

11. Disadvantages of AI Integration in Pharmacy

The integration of AI and ML in pharmaceutical research and development introduces a plethora of ethical and regulatory considerations that necessitate careful examination. Firstly, ethical implications arise concerning the responsible use of AI and ML algorithms in drug discovery and development [132]. These technologies may inadvertently perpetuate biases present in historical data, leading to disparities in healthcare outcomes and exacerbating inequities. Additionally, the opaque nature of complex machine learning models poses challenges to transparency and accountability, raising concerns regarding the reproducibility and interpretability of algorithmic decisions [133]. Moreover, regulatory frameworks governing AI-driven drug development must evolve to address novel challenges related to safety, efficacy, and data privacy. Striking a balance between innovation and regulatory compliance requires proactive engagement with regulatory agencies to ensure alignment with evolving standards and guidelines. Furthermore, safeguarding data privacy and confidentiality remains paramount in healthcare AI applications, necessitating robust measures to protect sensitive patient information and comply with stringent data protection regulations [134]. By embracing ethical principles and regulatory best practices, stakeholders can navigate the complex landscape of AI-driven pharmaceutical research while upholding patient welfare and societal trust.

Effects on the Environment

In addition to ethical and regulatory challenges, the integration of AI in the pharmaceutical industry has notable environmental implications that warrant attention. The computational intensity of AI algorithms, coupled with the infrastructure required for data processing and storage, contributes to significant energy consumption and carbon emissions. The proliferation of AI-driven technologies necessitates large-scale data centers and computing resources, which consume vast amounts of electricity, predominantly sourced from non-renewable energy sources [135]. Consequently, the carbon footprint associated with AI integration in the pharmaceutical sector raises concerns about environmental sustainability and climate change mitigation. Addressing these environmental challenges requires a multifaceted approach, encompassing energy-efficient algorithm design, adoption of renewable energy sources for computing infrastructure, and implementation of sustainable data center practices [136,137]. Various repercussions of AI/ML innovations have been summarized in Table 6. By prioritizing environmental sustainability alongside technological advancement, the pharmaceutical industry can mitigate its environmental impact and contribute to a more sustainable future.

Author Contributions

Priyanka Kandhare: Conceptualization, writing—original draft preparation; Mrunal Kurlekar: writing review and editing; Tanvi Deshpande: writing review and editing; Atmaram Pawar: visualization, supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare that there are no conflicts of interest.

Abbreviations

Sr. No.AbbreviationFull Form
1AIArtificial Intelligence
2MLMachine Learning
3RWDReal World Data
4NLPNatural Language Processing
5DLDeep Learning
6Ro5Rule of Five
7GEOGene Expression Omnibus
8TCGAThe Cancer Genome Atlas
9GWASGenome-Wide Association Studies
10QSARQuantitative Structure-Activity Relationship
11SMILESSimplified Molecular Input Line Entry System
12L-NetLigand Neural Network
13DFTDensity Functional Theory
14V-SYNTHESVirtual Synthon Hierarchical Enumeration Screening
15ADMETAbsorption Distribution Metabolism Elimination Toxicity
16GCNNGraph Convolutional Neural Network
17MRLMolecular Representation Learning
18CPConformal Prediction
19ACActivity Cliff
20ROCReceiver Operating Characteristic
21MDSMolecular Dynamics Simulation
22ACEAngiotensin Converting Enzyme
23mProMain Protease
24AIDDArtificial Intelligence-Driven Drug Design
25HINTHierarchical Interaction Network
26SPOTSequential Prediction Modeling of Clinical Trial Outcome
27RBMRisk-Based Management
28ADEAdverse Drug Event
29ADRAdverse Drug Reaction
30EHRElectronic Health Record
31FDAFood and Drug Administration
32PTMLPerturbation-Theory Machine Learning

References

  1. Olczak, J.; Pavlopoulos, J.; Prijs, J.; Ijpma, F.F.A.; Doornberg, J.N.; Lundström, C.; Hedlund, J.; Gordon, M. Presenting artificial intelligence, deep learning, and machine learning studies to clinicians and healthcare stakeholders: An introductory reference with a guideline and a Clinical AI Research (CAIR) checklist proposal. Acta Orthop. 2021, 92, 513–525. [Google Scholar] [CrossRef] [PubMed]
  2. Paul, D.; Sanap, G.; Shenoy, S.; Kalyane, D.; Kalia, K.; Tekade, R.K. Artificial intelligence in drug discovery and development. Drug Discov. Today 2021, 26, 80–93. [Google Scholar] [CrossRef]
  3. Gkouvas, N.; Gkouvas, N. Precision Medicine & Pharmacogenomics: Personalized Medication in Neuropsychiatric Disorders using AI and telepsychiatry. Eur. Psychiatry 2022, 65, S678. [Google Scholar] [CrossRef]
  4. Bello, B.K.; Bundey, Y.; Bhave, R.; Khotimchenko, M.; Baran, S.W.; Chakravarty, K.; Varshney, J. Integrating AI/ML Models for Patient Stratification Leveraging Omics Dataset and Clinical Biomarkers from COVID-19 Patients: A Promising Approach to Personalized Medicine. Int. J. Mol. Sci. 2023, 24, 6250. [Google Scholar] [CrossRef]
  5. Andrews, S.M. Emerging Role of Artificial Intelligence and Machine learning in precision medicine. Int. J. Eng. Technol. Manag. Sci. 2023, 7, 622–626. [Google Scholar] [CrossRef]
  6. Sanchez, P.; Sánchez, P.A.; Voisey, J.P.; Voisey, J.P.; Xia, T.; Xia, T.; Watson, H.I.; Watson, H.; O’Neil, A.Q.; O’Neil, A.Q.; et al. Causal machine learning for healthcare and precision medicine. R. Soc. Open Sci. 2022, 9, 220638. [Google Scholar] [CrossRef]
  7. Xu, Z.; Biswas, B.; Liu, L.; Amzal, B. AI/ML in Precision Medicine: A Look Beyond the Hype. Ther. Innov. Regul. Sci. 2023. [Google Scholar] [CrossRef]
  8. Han, Y.; Tao, J. Revolutionizing Pharma: Unveiling the AI and LLM Trends in the Pharmaceutical Industry. arXiv 2024, arXiv:2401.10273. [Google Scholar] [CrossRef]
  9. Terranova, N.; Renard, D.; Shahin, M.H.; Menon, S.; Cao, Y.; Hop, C.E.C.A.; Hayes, S.T.; Madrasi, K.; Stodtmann, S.; Tensfeldt, T.G.; et al. Artificial Intelligence for Quantitative Modeling in Drug Discovery and Development: An Innovation & Quality (IQ) Consortium Perspective on Use Cases and Best Practices. Clin. Pharmacol. Ther. 2023, 115, 658–672. [Google Scholar] [CrossRef]
  10. Knutson, C.; Bontha, M.; Bilbrey, J.A.; Kumar, N. Decoding the protein–ligand interactions using parallel graph neural networks. Sci. Rep. 2022, 12, 7624. [Google Scholar] [CrossRef]
  11. Shahzad, M.; Tahir, M.A.; Alhussein, M.; Mobin, A.; Malick, R.A.S.; Anwar, M.S. NeuPD—A Neural Network-Based Approach to Predict Antineoplastic Drug Response. Diagnostics 2023, 13, 2043. [Google Scholar] [CrossRef]
  12. Kolluri, S.; Lin, J.; Liu, R.; Zhang, Y.; Zhang, W. Machine Learning and Artificial Intelligence in Pharmaceutical Research and Development: A Review. AAPS J. 2022, 24, 19. [Google Scholar] [CrossRef] [PubMed]
  13. Yelne, S.; Chaudhary, M.; Dod, K.; Sayyad, A.; Sharma, R. Harnessing the Power of AI: A Comprehensive Review of Its Impact and Challenges in Nursing Science and Healthcare. Cureus 2023, 15, e49252. [Google Scholar] [CrossRef] [PubMed]
  14. Pinto-Coelho, L. How Artificial Intelligence Is Shaping Medical Imaging Technology: A Survey of Innovations and Applications. Bioengineering 2023, 10, 1435. [Google Scholar] [CrossRef] [PubMed]
  15. Liang, Y.; Sun, L.; Du, Z.; Yan, Z.; Wang, W. Mechanism Design and Optimization of a Haptic Master Manipulator for Laparoscopic Surgical Robots. IEEE Access 2019, 7, 147808–147824. [Google Scholar] [CrossRef]
  16. Kompa, B.; Hakim, J.B.; Palepu, A.; Kompa, K.G.; Smith, M.; Bain, P.A.; Woloszynek, S.; Painter, J.L.; Bate, A.; Beam, A.L. Artificial Intelligence Based on Machine Learning in Pharmacovigilance: A Scoping Review. Drug Saf. 2022, 45, 477–491. [Google Scholar] [CrossRef]
  17. FDA. Artificial Intelligence and Machine Learning (AI/ML) for Drug Development|FDA. Available online: https://www.fda.gov/science-research/science-and-research-special-topics/artificial-intelligence-and-machine-learning-aiml-drug-development (accessed on 4 January 2025).
  18. Bender, A.; Cortes-Ciriano, I. Artificial intelligence in drug discovery: What is realistic, what are illusions? Part 2: A discussion of chemical and biological data. Drug Discov. Today 2021, 26, 1040–1052. [Google Scholar] [CrossRef]
  19. Sarkar, C.; Das, B.; Rawat, V.S.; Wahlang, J.B.; Nongpiur, A.; Tiewsoh, I.; Lyngdoh, N.M.; Das, D.; Bidarolli, M.; Sony, H.T. Artificial Intelligence and Machine Learning Technology Driven Modern Drug Discovery and Development. Int. J. Mol. Sci. 2023, 24, 2026. [Google Scholar] [CrossRef]
  20. Wong, C.H.; Siah, K.W.; Lo, A.W. Estimation of clinical trial success rates and related parameters. Biostatistics 2019, 20, 273–286. [Google Scholar] [CrossRef]
  21. Gupta, R.; Srivastava, D.; Sahu, M.; Tiwari, S.; Ambasta, R.K.; Kumar, P. Artificial intelligence to deep learning: Machine intelligence approach for drug discovery. Mol. Divers. 2021, 25, 1315–1360. [Google Scholar] [CrossRef]
  22. Vamathevan, J.; Clark, D.; Czodrowski, P.; Dunham, I.; Ferran, E.; Lee, G.; Li, B.; Madabhushi, A.; Shah, P.; Spitzer, M.; et al. Applications of machine learning in drug discovery and development. Nat. Rev. Drug Discov. 2019, 18, 463–477. [Google Scholar] [CrossRef]
  23. Kalyane, D.; Sanap, G.; Paul, D.; Shenoy, S.; Anup, N.; Polaka, S.; Tambe, V.; Tekade, R.K. Artificial intelligence in the pharmaceutical sector: Current scene and future prospect. In The Future of Pharmaceutical Product Development and Research; Elsevier: Amsterdam, The Netherlands, 2020; pp. 73–107. ISBN 978-0-12-814455-8. [Google Scholar]
  24. Moffat, J.G.; Vincent, F.; Lee, J.A.; Eder, J.; Prunotto, M. Opportunities and challenges in phenotypic drug discovery: An industry perspective. Nat. Rev. Drug Discov. 2017, 16, 531–543. [Google Scholar] [CrossRef] [PubMed]
  25. Vatansever, S.; Schlessinger, A.; Wacker, D.; Kaniskan, H.Ü.; Jin, J.; Zhou, M.; Zhang, B. Artificial intelligence and machine learning-aided drug discovery in central nervous system diseases: State-of-the-arts and future directions. Med. Res. Rev. 2021, 41, 1427–1473. [Google Scholar] [CrossRef]
  26. Athar, M.; Lone, M.Y.; Jha, P.C. First protein drug target’s appraisal of lead-likeness descriptors to unfold the intervening chemical space. J. Mol. Graph. Model. 2017, 72, 272–282. [Google Scholar] [CrossRef] [PubMed]
  27. Lalhmangaihzuala, S.; Vanlaldinpuia, K.; Khiangte, V.; Laldinpuii, Z.; Liana, T.; Lalhriatpuia, C.; Pachuau, Z. Therapeutic applications of carbohydrate-based compounds: A sweet solution for medical advancement. Mol. Divers. 2024, 28, 4553–4579. [Google Scholar] [CrossRef]
  28. Markovic, M.; Ben-Shabat, S.; Aponick, A.; Zimmermann, E.M.; Dahan, A. Lipids and Lipid-Processing Pathways in Drug Delivery and Therapeutics. Int. J. Mol. Sci. 2020, 21, 3248. [Google Scholar] [CrossRef] [PubMed]
  29. Lo, Y.-C.; Ren, G.; Honda, H.; Davis, K.L. Artificial Intelligence-Based Drug Design and Discovery. In Cheminformatics and Its Applications; Stefaniu, A., Rasul, A., Hussain, G., Eds.; IntechOpen: London, UK, 2020; ISBN 978-1-83880-067-3. [Google Scholar]
  30. Buniello, A.; MacArthur, J.A.L.; Cerezo, M.; Harris, L.W.; Hayhurst, J.; Malangone, C.; McMahon, A.; Morales, J.; Mountjoy, E.; Sollis, E.; et al. The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res. 2019, 47, D1005–D1012. [Google Scholar] [CrossRef]
  31. Lau, A.; So, H.-C. Turning genome-wide association study findings into opportunities for drug repositioning. Comput. Struct. Biotechnol. J. 2020, 18, 1639–1650. [Google Scholar] [CrossRef]
  32. Valentini, G.; Paccanaro, A.; Caniza, H.; Romero, A.E.; Re, M. An extensive analysis of disease-gene associations using network integration and fast kernel-based gene prioritization methods. Artif. Intell. Med. 2014, 61, 63–78. [Google Scholar] [CrossRef]
  33. Senior, A.W.; Evans, R.; Jumper, J.; Kirkpatrick, J.; Sifre, L.; Green, T.; Qin, C.; Žídek, A.; Nelson, A.W.R.; Bridgland, A.; et al. Improved protein structure prediction using potentials from deep learning. Nature 2020, 577, 706–710. [Google Scholar] [CrossRef]
  34. Desai, D.; Kantliwala, S.V.; Vybhavi, J.; Ravi, R.; Patel, H.; Patel, J. Review of AlphaFold 3: Transformative Advances in Drug Design and Therapeutics. Cureus 2024, 16, e63646. [Google Scholar] [CrossRef] [PubMed]
  35. Kamya, P.; Ozerov, I.V.; Pun, F.W.; Tretina, K.; Fokina, T.; Chen, S.; Naumov, V.; Long, X.; Lin, S.; Korzinkin, M.; et al. PandaOmics: An AI-Driven Platform for Therapeutic Target and Biomarker Discovery. J. Chem. Inf. Model. 2024, 64, 3961–3969. [Google Scholar] [CrossRef]
  36. Chen, Y.; Elenee Argentinis, J.; Weber, G. IBM Watson: How Cognitive Computing Can Be Applied to Big Data Challenges in Life Sciences Research. Clin. Ther. 2016, 38, 688–701. [Google Scholar] [CrossRef] [PubMed]
  37. Makhouri, F.R.; Ghasemi, J.B. Combating Diseases with Computational Strategies Used for Drug Design and Discovery. Curr. Top. Med. Chem. 2019, 18, 2743–2773. [Google Scholar] [CrossRef] [PubMed]
  38. Jourdan, J.-P.; Bureau, R.; Rochais, C.; Dallemagne, P. Drug repositioning: A brief overview. J. Pharm. Pharmacol. 2020, 72, 1145–1151. [Google Scholar] [CrossRef]
  39. Hua, Y.; Dai, X.; Xu, Y.; Xing, G.; Liu, H.; Lu, T.; Chen, Y.; Zhang, Y. Drug repositioning: Progress and challenges in drug discovery for various diseases. Eur. J. Med. Chem. 2022, 234, 114239. [Google Scholar] [CrossRef]
  40. Zeng, X.; Zhu, S.; Lu, W.; Liu, Z.; Huang, J.; Zhou, Y.; Fang, J.; Huang, Y.; Guo, H.; Li, L.; et al. Target identification among known drugs by deep learning from heterogeneous networks. Chem. Sci. 2020, 11, 1775–1797. [Google Scholar] [CrossRef]
  41. Varikoti, R.A.; Schultz, K.J.; Kombala, C.J.; Kruel, A.; Brandvold, K.R.; Zhou, M.; Kumar, N. Integrated data-driven and experimental approaches to accelerate lead optimization targeting SARS-CoV-2 main protease. J. Comput. Aided Mol. Des. 2023, 37, 339–355. [Google Scholar] [CrossRef]
  42. Joshi, R.P.; Kumar, N. Artificial Intelligence for Autonomous Molecular Design: A Perspective. Molecules 2021, 26, 6761. [Google Scholar] [CrossRef]
  43. Arul Murugan, N.; Ruba Priya, G.; Narahari Sastry, G.; Markidis, S. Artificial intelligence in virtual screening: Models versus experiments. Drug Discov. Today 2022, 27, 1913–1923. [Google Scholar] [CrossRef]
  44. Li, Y.; Zhang, L.; Liu, Z. Multi-objective de novo drug design with conditional graph generative model. J. Cheminform. 2018, 10, 33. [Google Scholar] [CrossRef] [PubMed]
  45. Bian, Y.; Xie, X.-Q. Generative chemistry: Drug discovery with deep learning generative models. J. Mol. Model. 2021, 27, 71. [Google Scholar] [CrossRef]
  46. Li, Y.; Pei, J.; Lai, L. Structure-based de novo drug design using 3D deep generative models. Chem. Sci. 2021, 12, 13664–13675. [Google Scholar] [CrossRef] [PubMed]
  47. Atance, S.R.; Diez, J.V.; Engkvist, O.; Olsson, S.; Mercado, R. De Novo Drug Design Using Reinforcement Learning with Graph-Based Deep Generative Models. J. Chem. Inf. Model. 2022, 62, 4863–4872. [Google Scholar] [CrossRef]
  48. Keith, J.A.; Vassilev-Galindo, V.; Cheng, B.; Chmiela, S.; Gastegger, M.; Müller, K.-R.; Tkatchenko, A. Combining Machine Learning and Computational Chemistry for Predictive Insights Into Chemical Systems. Chem. Rev. 2021, 121, 9816–9872. [Google Scholar] [CrossRef]
  49. Shi, Y.-F.; Yang, Z.-X.; Ma, S.; Kang, P.-L.; Shang, C.; Hu, P.; Liu, Z.-P. Machine Learning for Chemistry: Basics and Applications. Engineering 2023, 27, 70–83. [Google Scholar] [CrossRef]
  50. Glen, R.C. Computational chemistry and cheminformatics: An essay on the future. J. Comput. Aided Mol. Des. 2012, 26, 47–49. [Google Scholar] [CrossRef] [PubMed]
  51. Gryniukova, A.; Kaiser, F.; Myziuk, I.; Alieksieieva, D.; Leberecht, C.; Heym, P.P.; Tarkhanova, O.O.; Moroz, Y.S.; Borysko, P.; Haupt, V.J. AI-Powered Virtual Screening of Large Compound Libraries Leads to the Discovery of Novel Inhibitors of Sirtuin-1. J. Med. Chem. 2023, 66, 10241–10251. [Google Scholar] [CrossRef]
  52. Sotriffer, C. (Ed.) Virtual Screening: Principles, Challenges, and Practical Guidelines, 1st ed.; Methods and Principles in Medicinal Chemistry; Wiley: Hoboken, NJ, USA, 2011; ISBN 978-3-527-32636-5. [Google Scholar]
  53. Murugan, N.A.; Podobas, A.; Gadioli, D.; Vitali, E.; Palermo, G.; Markidis, S. A Review on Parallel Virtual Screening Softwares for High-Performance Computers. Pharmaceuticals 2022, 15, 63. [Google Scholar] [CrossRef]
  54. Lavecchia, A.; Giovanni, C. Virtual Screening Strategies in Drug Discovery: A Critical Review. Curr. Med. Chem. 2013, 20, 2839–2860. [Google Scholar] [CrossRef]
  55. Deane, C.; Mokaya, M. A virtual drug-screening approach to conquer huge chemical libraries. Nature 2022, 601, 322–323. [Google Scholar] [CrossRef]
  56. Jung, S.; Vatheuer, H.; Czodrowski, P. VSFlow: An open-source ligand-based virtual screening tool. J. Cheminform. 2023, 15, 40. [Google Scholar] [CrossRef] [PubMed]
  57. Zhou, G.; Rusnac, D.-V.; Park, H.; Canzani, D.; Nguyen, H.M.; Stewart, L.; Bush, M.F.; Nguyen, P.T.; Wulff, H.; Yarov-Yarovoy, V.; et al. An artificial intelligence accelerated virtual screening platform for drug discovery. Nat. Commun. 2024, 15, 7761. [Google Scholar] [CrossRef] [PubMed]
  58. Buvailo, A. AI Takes on DNA-encoded Chemical Libraries. Available online: https://enamine.net/blog/ai-takes-on-dna-encoded-chemical-libraries (accessed on 4 January 2025).
  59. Golbraikh, A.; Wang, X.S.; Zhu, H.; Tropsha, A. Predictive QSAR Modeling: Methods and Applications in Drug Discovery and Chemical Risk Assessment. In Handbook of Computational Chemistry; Leszczynski, J., Kaczmarek-Kedziera, A., Puzyn, T.G., Papadopoulos, M., Reis, H., Shukla, M.K., Eds.; Springer International Publishing: Cham, Switzerland, 2017; pp. 2303–2340. ISBN 978-3-319-27281-8. [Google Scholar]
  60. Xu, Y.; Liaw, A.; Sheridan, R.P.; Svetnik, V. Development and Evaluation of Conformal Prediction Methods for QSAR. arXiv 2023, arXiv:2304.00970. [Google Scholar]
  61. Kwon, S.; Bae, H.; Jo, J.; Yoon, S. Comprehensive ensemble in QSAR prediction for drug discovery. BMC Bioinform. 2019, 20, 521. [Google Scholar] [CrossRef]
  62. Gao, Z.; Ji, X.; Zhao, G.; Wang, H.; Zheng, H.; Ke, G.; Zhang, L. Uni-QSAR: An Auto-ML Tool for Molecular Property Prediction. arXiv 2023, arXiv:2304.12239. [Google Scholar]
  63. Ambure, P.; Halder, A.K.; González Díaz, H.; Cordeiro, M.N.D.S. QSAR-Co: An Open Source Software for Developing Robust Multitasking or Multitarget Classification-Based QSAR Models. J. Chem. Inf. Model. 2019, 59, 2538–2544. [Google Scholar] [CrossRef]
  64. Vukovic, K.; Gadaleta, D.; Benfenati, E. Methodology of aiQSAR: A group-specific approach to QSAR modelling. J. Cheminform. 2019, 11, 27. [Google Scholar] [CrossRef]
  65. Dablander, M.; Hanser, T.; Lambiotte, R.; Morris, G.M. Exploring QSAR models for activity-cliff prediction. J. Cheminform. 2023, 15, 47. [Google Scholar] [CrossRef]
  66. Al-Karmalawy, A.A.; Dahab, M.A.; Metwaly, A.M.; Elhady, S.S.; Elkaeed, E.B.; Eissa, I.H.; Darwish, K.M. Molecular Docking and Dynamics Simulation Revealed the Potential Inhibitory Activity of ACEIs Against SARS-CoV-2 Targeting the hACE2 Receptor. Front. Chem. 2021, 9, 661230. [Google Scholar] [CrossRef]
  67. Crampon, K.; Giorkallos, A.; Deldossi, M.; Baud, S.; Steffenel, L.A. Machine-learning methods for ligand–protein molecular docking. Drug Discov. Today 2022, 27, 151–164. [Google Scholar] [CrossRef] [PubMed]
  68. Vidal-Limon, A.; Aguilar-Toalá, J.E.; Liceaga, A.M. Integration of Molecular Docking Analysis and Molecular Dynamics Simulations for Studying Food Proteins and Bioactive Peptides. J. Agric. Food Chem. 2022, 70, 934–943. [Google Scholar] [CrossRef]
  69. Niazi, S. The Coming of Age of AI/ML in Drug Discovery, Development, Clinical Testing, and Manufacturing: The FDA Perspectives. Drug Des. Devel. Ther. 2023, 17, 2691–2725. [Google Scholar] [CrossRef] [PubMed]
  70. Rafiq, H.; Hu, J.; Hakami, M.A.; Hazazi, A.; Alamri, M.A.; Alkhatabi, H.A.; Mahmood, A.; Alotaibi, B.S.; Wadood, A.; Huang, X. Identification of novel STAT3 inhibitors for liver fibrosis, using pharmacophore-based virtual screening, molecular docking, and biomolecular dynamics simulations. Sci. Rep. 2023, 13, 20147. [Google Scholar] [CrossRef]
  71. Mustali, J.; Yasuda, I.; Hirano, Y.; Yasuoka, K.; Gautieri, A.; Arai, N. Unsupervised deep learning for molecular dynamics simulations: A novel analysis of protein–ligand interactions in SARS-CoV-2 M pro. RSC Adv. 2023, 13, 34249–34261. [Google Scholar] [CrossRef]
  72. Zhang, Y. An In-depth Summary of Recent Artificial Intelligence Applications in Drug Design. arXiv 2021, arXiv:2110.05478. [Google Scholar]
  73. Jones, J.; Clark, R.D.; Lawless, M.S.; Miller, D.W.; Waldman, M. The AI-driven Drug Design (AIDD) platform: An interactive multi-parameter optimization system integrating molecular evolution with physiologically based pharmacokinetic simulations. J. Comput. Aided Mol. Des. 2024, 38, 14. [Google Scholar] [CrossRef]
  74. Batool, M.; Ahmad, B.; Choi, S. A Structure-Based Drug Discovery Paradigm. Int. J. Mol. Sci. 2019, 20, 2783. [Google Scholar] [CrossRef]
  75. Bai, Q.; Liu, S.; Tian, Y.; Xu, T.; Banegas-Luna, A.J.; Pérez-Sánchez, H.; Huang, J.; Liu, H.; Yao, X. Application advances of deep learning methods for de novo drug design and molecular dynamics simulation. WIREs Comput. Mol. Sci. 2022, 12, e1581. [Google Scholar] [CrossRef]
  76. Ismail, A.; Al-Zoubi, T.; El Naqa, I.; Saeed, H. The role of artificial intelligence in hastening time to recruitment in clinical trials. BJR|Open 2023, 5, 20220023. [Google Scholar] [CrossRef]
  77. Hutson, M. How AI is being used to accelerate clinical trials. Nature 2024, 627, S2–S5. [Google Scholar] [CrossRef] [PubMed]
  78. Askin, S.; Burkhalter, D.; Calado, G.; El Dakrouni, S. Artificial Intelligence Applied to clinical trials: Opportunities and challenges. Health Technol. 2023, 13, 203–213. [Google Scholar] [CrossRef]
  79. Fu, T.; Huang, K.; Xiao, C.; Glass, L.M.; Sun, J. HINT: Hierarchical interaction network for clinical-trial-outcome predictions. Patterns 2022, 3, 100445. [Google Scholar] [CrossRef] [PubMed]
  80. Johnson, K.B.; Wei, W.; Weeraratne, D.; Frisse, M.E.; Misulis, K.; Rhee, K.; Zhao, J.; Snowdon, J.L. Precision Medicine, AI, and the Future of Personalized Health Care. Clin. Transl. Sci. 2021, 14, 86–93. [Google Scholar] [CrossRef]
  81. Bohr, A.; Memarzadeh, K. The rise of artificial intelligence in healthcare applications. In Artificial Intelligence in Healthcare; Elsevier: Amsterdam, The Netherlands, 2020; pp. 25–60. ISBN 978-0-12-818438-7. [Google Scholar]
  82. Herrera-Ibatá, D.M. Machine Learning and Perturbation Theory Machine Learning (PTML) in Medicinal Chemistry, Biotechnology, and Nanotechnology. Curr. Top. Med. Chem. 2021, 21, 649–660. [Google Scholar] [CrossRef] [PubMed]
  83. Kleandrova, V.V.; Scotti, M.T.; Scotti, L.; Speck-Planche, A. Multi-target Drug Discovery via PTML Modeling: Applications to the Design of Virtual Dual Inhibitors of CDK4 and HER2. Curr. Top. Med. Chem. 2021, 21, 661–675. [Google Scholar] [CrossRef]
  84. Speck-Planche, A.; Kleandrova, V.V. Multi-Condition QSAR Model for the Virtual Design of Chemicals with Dual Pan-Antiviral and Anti-Cytokine Storm Profiles. ACS Omega 2022, 7, 32119–32130. [Google Scholar] [CrossRef]
  85. Kleandrova, V.V.; Cordeiro, M.N.D.; Speck-Planche, A. Perturbation Theory Machine Learning Model for Phenotypic Early Antineoplastic Drug Discovery: Design of Virtual Anti-Lung-Cancer Agents. Appl. Sci. 2024, 14, 9344. [Google Scholar] [CrossRef]
  86. Kleandrova, V.V.; Speck-Planche, A. PTML Modeling for Pancreatic Cancer Research: In Silico Design of Simultaneous Multi-Protein and Multi-Cell Inhibitors. Biomedicines 2022, 10, 491. [Google Scholar] [CrossRef]
  87. Cabrera-Andrade, A.; López-Cortés, A.; Munteanu, C.R.; Pazos, A.; Pérez-Castillo, Y.; Tejera, E.; Arrasate, S.; Gonzalez-Diaz, H. Perturbation-Theory Machine Learning (PTML) Multilabel Model of the ChEMBL Dataset of Preclinical Assays for Antisarcoma Compounds. ACS Omega 2020, 5, 27211–27220. [Google Scholar] [CrossRef]
  88. Kleandrova, V.V.; Scotti, M.T.; Scotti, L.; Nayarisseri, A.; Speck-Planche, A. Cell-based multi-target QSAR model for design of virtual versatile inhibitors of liver cancer cell lines. Sar Qsar Environ. Res. 2020, 31, 815–836. [Google Scholar] [CrossRef]
  89. Kleandrova, V.V.; Cordeiro, M.N.D.; Speck-Planche, A. In Silico Approach for Antibacterial Discovery: PTML Modeling of Virtual Multi-Strain Inhibitors Against Staphylococcus aureus. Pharmaceuticals 2025, 18, 196. [Google Scholar] [CrossRef] [PubMed]
  90. Velásquez-López, Y.; Ruiz-Escudero, A.; Arrasate, S.; González-Díaz, H. Implementation of IFPTML Computational Models in Drug Discovery Against Flaviviridae Family. J. Chem. Inf. Model. 2024, 64, 1841–1852. [Google Scholar] [CrossRef] [PubMed]
  91. Kleandrova, V.V.; Scotti, M.T.; Speck-Planche, A. Computational Drug Repurposing for Antituberculosis Therapy: Discovery of Multi-Strain Inhibitors. Antibiotics 2021, 10, 1005. [Google Scholar] [CrossRef] [PubMed]
  92. Santiago, C.; Ortega-Tenezaca, B.; Barbolla, I.; Fundora-Ortiz, B.; Arrasate, S.; Dea-Ayuela, M.A.; González-Díaz, H.; Sotomayor, N.; Lete, E. Prediction of Antileishmanial Compounds: General Model, Preparation, and Evaluation of 2-Acylpyrrole Derivatives. J. Chem. Inf. Model. 2022, 62, 3928–3940. [Google Scholar] [CrossRef]
  93. Kleandrova, V.V.; Scotti, L.; Bezerra Mendonça, F.J., Jr.; Muratov, E.; Scotti, M.T.; Speck-Planche, A. QSAR Modeling for Multi-Target Drug Discovery: Designing Simultaneous Inhibitors of Proteins in Diverse Pathogenic Parasites. Front. Chem. 2021, 9, 634663. [Google Scholar] [CrossRef]
  94. Speck-Planche, A.; Kleandrova, V.V. Chapter 16: Demystifying Artificial Neural Networks as Generators of New Chemical Knowledge: Antimalarial Drug Discovery as a Case Study. In Machine Learning in Chemistry; The Royal Society of Chemistry: Cambridge, UK, 2020; pp. 398–423. [Google Scholar] [CrossRef]
  95. Kleandrova, V.V.; Cordeiro, M.N.D.; Speck-Planche, A. Perturbation-theory machine learning for mood disorders: Virtual design of dual inhibitors of NET and SERT proteins. BMC Chem. 2025, 19, 2. [Google Scholar] [CrossRef]
  96. Speck-Planche, A.; Kleandrova, V.V.; Scotti, M.T. In Silico Drug Repurposing for Anti-Inflammatory Therapy: Virtual Search for Dual Inhibitors of Caspase-1 and TNF-Alpha. Biomolecules 2021, 11, 1832. [Google Scholar] [CrossRef]
  97. Kleandrova, V.V.; Speck-Planche, A. PTML Modeling for Alzheimer’s Disease: Design and Prediction of Virtual Multi-Target Inhibitors of GSK3B, HDAC1, and HDAC6. Curr. Top. Med. Chem. 2020, 20, 1661–1676. [Google Scholar] [CrossRef]
  98. Speck-Planche, A.; Scotti, M.T. BET bromodomain inhibitors: Fragment-based in silico design using multi-target QSAR models. Mol. Divers. 2019, 23, 555–572. [Google Scholar] [CrossRef]
  99. Salas, M.; Petracek, J.; Yalamanchili, P.; Aimer, O.; Kasthuril, D.; Dhingra, S.; Junaid, T.; Bostic, T. The Use of Artificial Intelligence in Pharmacovigilance: A Systematic Review of the Literature. Pharm. Med. 2022, 36, 295–306. [Google Scholar] [CrossRef] [PubMed]
  100. Huysentruyt, K.; Kjoersvik, O.; Dobracki, P.; Savage, E.; Mishalov, E.; Cherry, M.; Leonard, E.; Taylor, R.; Patel, B.; Abatemarco, D. Validating Intelligent Automation Systems in Pharmacovigilance: Insights from Good Manufacturing Practices. Drug Saf. 2021, 44, 261–272. [Google Scholar] [CrossRef] [PubMed]
  101. Macrae, C. Governing the safety of artificial intelligence in healthcare. BMJ Qual. Saf. 2019, 28, 495–498. [Google Scholar] [CrossRef]
  102. Basile, A.O.; Yahi, A.; Tatonetti, N.P. Artificial Intelligence for Drug Toxicity and Safety. Trends Pharmacol. Sci. 2019, 40, 624–635. [Google Scholar] [CrossRef] [PubMed]
  103. Bates, D.W.; Levine, D.; Syrowatka, A.; Kuznetsova, M.; Craig, K.J.T.; Rui, A.; Jackson, G.P.; Rhee, K. The potential of artificial intelligence to improve patient safety: A scoping review. Npj Digit. Med. 2021, 4, 54. [Google Scholar] [CrossRef]
  104. Shamayleh, A.; Shamayleh, A.; Awad, M.; Awad, M.; Farhat, J.; Farhat, J.; Farhat, J. IoT Based Predictive Maintenance Management of Medical Equipment. J. Med. Syst. 2020, 44, 72. [Google Scholar] [CrossRef]
  105. Krishnamurthy, R.; Krishnamurthy, R.; Mummudi, N.; Mummudi, N.; Goda, J.S.; Goda, J.S.; Chopra, S.; Chopra, S.; Heijmen, B.; Heijmen, B.; et al. Using Artificial Intelligence for Optimization of the Processes and Resource Utilization in Radiotherapy. JCO Glob. Oncol. 2022, 8, e2100393. [Google Scholar] [CrossRef]
  106. Gao, Y.; Gao, H.; Han, X.; Yao, F. Vaccine supply chain coordination using blockchain and artificial intelligence technologies. Comput. Ind. Eng. 2023, 175, 108885. [Google Scholar] [CrossRef]
  107. Duarte, J.M.; Veiga, F.; Mascarenhas-Melo, F. Exploiting Pharma 4.0 Technologies in the Non-Biological Complex Drugs Manufacturing: Innovations and Implications. Pharmaceutics 2023, 15, 2545. [Google Scholar] [CrossRef]
  108. Abisha, D.; Varshini, S.; Thejaswini, A.; Sineka, P. Blockchain-Enabled Pharmacy Supply Chain Management: Ensuring Transparency and Efficiency. In Proceedings of the 2023 International Conference on Sustainable Communication Networks and Application (ICSCNA), Theni, India, 15–17 November 2023. [CrossRef]
  109. Santosh, K.C.; Gaur, L. AI in Precision Medicine. Artif. Intell. Mach. Learn. Public Healthc. 2021, 41–47. [Google Scholar] [CrossRef]
  110. DeGroat, W.; Abdelhalim, H.; Patel, K.; Mendhe, D.; Zeeshan, S.; Ahmed, Z. Discovering biomarkers associated and predicting cardiovascular disease with high accuracy using a novel nexus of machine learning techniques for precision medicine. Sci. Rep. 2024, 14, 1. [Google Scholar] [CrossRef]
  111. Restrepo, J.C.; Dueñas, D.; Corredor, Z.; Liscano, Y. Advances in Genomic Data and Biomarkers: Revolutionizing NSCLC Diagnosis and Treatment. Cancers 2023, 15, 3474. [Google Scholar] [CrossRef] [PubMed]
  112. Terranova, N.; Venkatakrishnan, K. Machine Learning in Modeling Disease Trajectory and Treatment Outcomes: An Emerging Enabler for Model-Informed Precision Medicine. Clin. Pharmacol. Ther. 2024, 115, 720–726. [Google Scholar] [CrossRef] [PubMed]
  113. Napitupulu, P.A. Ethical Dilemmas in the Use of Artificial Intelligence in Breast Cancer Diagnosis and Treatment (Addressing Issues of Bias, Transferability, and Patient Trust in Breast Cancer AI). West Sci. Law Hum. Rights 2023, 1, 4. [Google Scholar] [CrossRef]
  114. Sharma, A.K. A study on the applicability of AI in Pharmaceutical Industry. In Proceedings of the 2022 1st International Conference on Computational Science and Technology (ICCST), Chennai, India, 9–10 November 2022. [Google Scholar] [CrossRef]
  115. Nagaprasad, S.; Padmaja, D.L.; Qureshi, Y.; Bangare, S.L.; Mishra, M.; Mazumdar, B.D. Investigating the Impact of Machine Learning in Pharmaceutical Industry. J. Pharm. Res. 2021, 33, 6–14. [Google Scholar] [CrossRef]
  116. Pedraza, D.H.; de la Cruz, M.L.; Nettleton, D.F.; Solà, P.A. Integrating Artificial Intelligence and Big Data in Pharmaceutical Development: Ethical Considerations and Legislative Frameworks. Available online: https://www.eternalproject.eu/downloads/publications/ai_and_big_data_in_pharma_dev.pdf (accessed on 3 February 2025).
  117. Lekadir, K.; Frangi, A.F.; Porras, A.R.; Glocker, B.; Cintas, C.; Langlotz, C.P.; Weicken, E.; Asselbergs, F.W.; Prior, F.; Collins, G.S.; et al. FUTURE-AI: International consensus guideline for trustworthy and deployable artificial intelligence in healthcare. arXiv 2023, arXiv:2309.12325. [Google Scholar] [CrossRef]
  118. Pashkov, V.; Soloviov, O. Legal implementation of blockchain technology in pharmacy. SHS Web Conf. 2019, 68, 01027. [Google Scholar] [CrossRef]
  119. Beauxis-Aussalet, E.; Behrisch, M.; Borgo, R.; Chau, D.H.; Collins, C.; Ebert, D.; El-Assady, M.; Endert, A.; Keim, D.A.; Kohlhammer, J.; et al. The Role of Interactive Visualization in Fostering Trust in AI. IEEE Comput. Graph. Appl. 2021, 41, 7–12. [Google Scholar] [CrossRef]
  120. Busuioc, M. Accountable Artificial Intelligence: Holding Algorithms to Account. Public Adm. Rev. 2020, 81, 825–836. [Google Scholar] [CrossRef]
  121. Saeed, G.; Kohler, J.C.; Cuomo, R.E.; Mackey, T.K. A systematic review of digital technology and innovation and its potential to address anti-corruption, transparency, and accountability in the pharmaceutical supply chain. Expert Opin. Drug Saf. 2022, 21, 1061–1088. [Google Scholar] [CrossRef]
  122. Frasca, M.; La Torre, D.; Pravettoni, G.; Cutica, I. Explainable and interpretable artificial intelligence in medicine: A systematic bibliometric review. Discov. Artif. Intell. 2024, 4, 15. [Google Scholar] [CrossRef]
  123. Goswami, R.; Yadav, S.; Kumar, V. Explainable AI in healthcare: A theoretical overview of interpretable models for medical diagnosis. Pharma Innov. 2019, 8, 29–33. [Google Scholar] [CrossRef]
  124. Sandeep Reddy Explainability and artificial intelligence in medicine. Lancet Digit. Health 2022, 4, e214–e215. [CrossRef] [PubMed]
  125. Bhosale, A.; Sawant, A.; Kamble, T.; Chougule, N. Exploring the Role of Artificial Intelligence in Modernizing Quality Assurance and Quality Control in the Pharmaceutical Sector. Int. J. Pharm. Sci. 2024, 2, 522–529. [Google Scholar] [CrossRef]
  126. Singh, D.; Chaddah, J.K. A Study On Application of Blockchain Technology to Control Counterfeit Drugs, Enhance Data Privacy and Improve Distribution in Online Pharmacy. Asia Pac. J. Health Manag. 2021, 16, 59–66. [Google Scholar] [CrossRef]
  127. Mahesh, A.B.; Reche, A. A New Era of Dental Care: Harnessing Artificial Intelligence for Better Diagnosis and Treatment. Cureus 2023, 15, e49319. [Google Scholar] [CrossRef]
  128. Danishuddin; Jamal, M.S.; Song, K.-S.; Lee, K.-W.; Kim, J.-J.; Park, Y.-M. Revolutionizing Drug Targeting Strategies: Integrating Artificial Intelligence and Structure-Based Methods in PROTAC Development. Pharmaceuticals 2023, 16, 1649. [Google Scholar] [CrossRef]
  129. Frascarelli, C.; Bonizzi, G.; Musico, C.R.; Mane, E.; Cassi, C.; Guerini-Rocco, E.; Farina, A.; Scarpa, A.; Lawlor, R.T.; Bonetti, L.R.; et al. Revolutionizing Cancer Research: The Impact of Artificial Intelligence in Digital Biobanking. J. Pers. Med. 2023, 13, 1390. [Google Scholar] [CrossRef]
  130. McComb, M.; McComb, M.; Bies, R.R.; Bies, R.R.; Ramanathan, M.; Ramanathan, M. Machine learning in pharmacometrics: Opportunities and challenges. Br. J. Clin. Pharmacol. 2021, 88, 1482–1499. [Google Scholar] [CrossRef]
  131. Dave, M.; Patel, N. Artificial intelligence in healthcare and education. Br. Dent. J. 2023, 234, 761–764. [Google Scholar] [CrossRef]
  132. Alshuhri, M.S.; Al-Musawi, S.G.; Al-Alwany, A.A.; Uinarni, H.; Rasulova, I.; Rodrigues, P.; Alkhafaji, A.T.; Alshanberi, A.M.; Alawadi, A.H.; Abbas, A.H. Artificial intelligence in cancer diagnosis: Opportunities and challenges. Pathol. Res. Pract. 2024, 253, 154996. [Google Scholar] [CrossRef] [PubMed]
  133. Jahn, S.W.; Plass, M.; Moinfar, F. Digital Pathology: Advantages, Limitations and Emerging Perspectives. J. Clin. Med. 2020, 9, 3697. [Google Scholar] [CrossRef]
  134. Chen, P.; Gao, J.; Ji, Z.; Liang, H.; Peng, Y. Do Artificial Intelligence Applications Affect Carbon Emission Performance?—Evidence from Panel Data Analysis of Chinese Cities. Energies 2022, 15, 5730. [Google Scholar] [CrossRef]
  135. Farchi, F.; Farchi, C.; Touzi, B.; Mabrouki, C. A Comparative Study on AI-Based Algorithms for Cost Prediction in Pharmaceutical Transport Logistics. Acadlore Trans. AI Mach. Learn. 2023, 2, 129–141. [Google Scholar] [CrossRef]
  136. Zhong, J.; Zhong, Y.; Han, M.; Yang, T.; Zhang, Q. The impact of AI on carbon emissions: Evidence from 66 countries. Appl. Econ. 2023, 56, 2975–2989. [Google Scholar] [CrossRef]
  137. Dubey, A.; Yadav, A. New Era’s of Artificial Intelligence in Pharmaceutical Industries. Asian J. Pharm. Res. Dev. 2024, 12, 71–76. [Google Scholar] [CrossRef]
  138. Sangeetha, M.; Hoti, A.; Bansal, R.; Hasan, M.F.; Gajjar, K.; Srivastava, K. Facilitating artificial intelligence supply chain analytics through finance management during the pandemic crises. Mater. Today Proc. 2021, 56, 2092–2095. [Google Scholar] [CrossRef]
  139. Paschek, D.; Luminosu, C.T.; Draghici, A. Automated business process management—In times of digital transformation using machine learning or artificial intelligence. MATEC Web Conf. 2017, 121, 04007. [Google Scholar] [CrossRef]
  140. Sommerfeld, S.; Strube, J. Challenges in biotechnology production—Generic processes and process optimization for monoclonal antibodies. Chem. Eng. Process. 2005, 44, 1123–1137. [Google Scholar] [CrossRef]
  141. Kumar, R.; Sood, P. Uses of AI in Field of Radiology- What is State of Doctor & Pateints Communication in Different Disease for Diagnosis Purpose. J. Res. Appl. Sci. Biotechnol. 2023, 2, 51–60. [Google Scholar] [CrossRef]
  142. Sharma, D.K.; Chatterjee, M.; Kaur, G.; Vavilala, S. Deep learning applications for disease diagnosis. In Deep Learning for Medical Applications with Unique Data; Academic Press: Cambridge, MA, USA, 2022. [Google Scholar] [CrossRef]
  143. Samala, A.; Rawas, S. Generative AI as Virtual Healthcare Assistant for Enhancing Patient Care Quality. Int. J. Online Biomed. Eng. 2024, 20, 174–187. [Google Scholar] [CrossRef]
  144. Stypińska, J.; Franke, A. AI revolution in healthcare and medicine and the (re-)emergence of inequalities and disadvantages for ageing population. Front. Sociol. 2023, 7, 1038854. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Types of machine learning algorithms.
Figure 1. Types of machine learning algorithms.
Ddc 04 00009 g001
Figure 2. Various applications of AI/ML in drug discovery.
Figure 2. Various applications of AI/ML in drug discovery.
Ddc 04 00009 g002
Figure 3. Applications of AI across the drug discovery continuum.
Figure 3. Applications of AI across the drug discovery continuum.
Ddc 04 00009 g003
Figure 4. Integration of AI/ML in QSAR workflow.
Figure 4. Integration of AI/ML in QSAR workflow.
Ddc 04 00009 g004
Figure 5. Various AI tools and their uses across the clinical trial process.
Figure 5. Various AI tools and their uses across the clinical trial process.
Ddc 04 00009 g005
Figure 6. Applications of blockchain in pharmaceutical industry.
Figure 6. Applications of blockchain in pharmaceutical industry.
Ddc 04 00009 g006
Table 2. Applications of PTML across various fields in drug discovery.
Table 2. Applications of PTML across various fields in drug discovery.
Application AreaStudy TitleKey FindingsReferences
Anticancer ResearchPTML for phenotypic early antineoplastic drug discoveryDesigned virtual anti-lung-cancer agents with optimized multi-target activity[86]
PTML modeling for pancreatic cancer researchIdentified simultaneous multi-protein and multi-cell inhibitors[87]
Multilabel model of the ChEMBL dataset of preclinical assays for antisarcoma compoundsEnabled prediction of multi-condition anticancer efficacy[88]
Cell-based multi-target QSAR modelDesigned virtual versatile inhibitors for liver cancer cell lines[89]
Antimicrobial AgentsIn Silico Approach for Antibacterial DiscoveryDesigned inhibitors against multi-strain S. aureus infections[90]
Implementation of IFPTML computational modelsDrug discovery against Flaviviridae family[91]
Multi-Condition QSAR ModelDesigned chemicals with dual pan-antiviral and anti-cytokine storm profiles[85]
Computational Drug Repurposing for TuberculosisDiscovered multi-strain inhibitors for tuberculosis therapy[92]
Prediction of Antileishmanial CompoundsDesigned and evaluated 2-acylpyrrole derivatives[93]
QSAR Modeling for Multi-Target Drug DiscoveryDesigned inhibitors for diverse pathogenic parasites[94]
Demystifying Artificial Neural Networks in Drug DiscoveryApplied AI for antimalarial compound discovery[95]
Dual-Target/Multi-Target InhibitorsPTML for Mood DisordersDesigned inhibitors targeting NET and SERT proteins[96]
In Silico Drug Repurposing for Anti-Inflammatory TherapyIdentified dual inhibitors of caspase-1 and TNF-alpha[97]
Multi-Target Drug Discovery via PTMLDesigned virtual dual inhibitors of CDK4 and HER2[84]
PTML Modeling for Alzheimer’s DiseaseDesigned multi-target inhibitors for GSK3B, HDAC1, and HDAC6[98]
BET Bromodomain InhibitorsDesigned inhibitors using fragment-based QSAR modeling[99]
Table 3. Specific parameters relevant to precision medicine along with respective ML models employed for them.
Table 3. Specific parameters relevant to precision medicine along with respective ML models employed for them.
Specific ParameterDescriptionRelevance to Precision MedicineML Model UsedReferences
Patient demographicsAge, gender, race, ethnicityDisease risk, treatment responseDecision Trees, Random Forest, Logistic Regression[4,13,63]
Clinical historyPast medical history, family history, lifestyle factorsDisease risk, progressionDecision Trees, Random Forest, Logistic Regression[4,13,63]
Genetic dataGenomic, epigenetic, transcriptomic dataDisease risk, progression, treatment responseRandom Forest, Support Vector Machine, Deep Learning[4,13,63]
Imaging dataRadiologic, pathologic imagesDisease severity, progressionConvolutional Neural Networks (CNN), Deep Learning[4,13,63]
Laboratory dataBlood tests, urine tests, other laboratory measuresDisease status, treatment responseDecision Trees, Random Forest, Logistic Regression[4,13,63]
Environmental dataEnvironmental exposuresDisease risk, progressionDecision Trees, Random Forest, Logistic Regression[4,13,63]
Therapeutic indexRatio of therapeutic to toxic doseDosing decisionsDecision Trees, Random Forest, Logistic Regression[4,13,63]
PK/PD variabilityVariability in drug absorption, distribution, metabolism, excretionTreatment responseDecision Trees, Random Forest, Logistic Regression[4,13,63]
BiomarkersMeasurable biological markersGuiding individualized dosingRandom Forest, Support Vector Machine, Deep Learning[4,13,63]
Disease severity and progressionTumor size, stage, other measures of disease severity and progressionTreatment responseDecision Trees, Random Forest, Logistic Regression[4,13,63]
PharmacoeconomicsCost of drug therapyTreatment decisions, resource allocationDecision Trees, Random Forest, Logistic Regression[4,13,63]
Dose-exposure and exposure-response relationshipsRelationship between drug dose, exposure, and responseInforming precision dosing strategiesDecision Trees, Random Forest, Logistic Regression[4,13,63]
Real-world patient gapIncongruity between study patients and patients in the real worldGeneralizability of clinical trial resultsDecision Trees, Random Forest, Logistic Regression[4,13,63]
Personalized treatment plansTailored treatment plans based on patient-specific factorsImproved patient outcomes, reduced healthcare costsDecision Trees, Random Forest, Logistic Regression[4,13,63]
Predictive analyticsPredicting patient outcomes based on historical dataImproved patient outcomes, reduced healthcare costsRandom Forest, Support Vector Machine, Deep Learning[4,13,63]
Real-time monitoringContinuous monitoring of patient health dataImproved patient outcomes, reduced healthcare costsDeep Learning, Recurrent Neural Networks (RNN), Long Short-Term Memory (LSTM)[4,13,63]
Table 4. Current applications of ML models by various companies in precision medicine.
Table 4. Current applications of ML models by various companies in precision medicine.
ApplicationLanguage ModelTypeCompanyReference
Predictive MaintenanceRandom ForestSupervised LearningGE Healthcarehttps://www.ge.com/digital/predix-asset-performance-management, accessed on 4 January 2025
Production Process OptimizationNeural NetworkSupervised LearningMerckhttps://www.merckgroup.com/en/research/open-innovation/merck-digital-science.html, accessed on 4 January 2025
Demand Forecasting and Inventory ManagementLong Short-Term Memory (LSTM)Sequence PredictionPfizerhttps://www.pfizer.com/research/science/ai, accessed on 4 January 2025
Supply Chain OptimizationSupport Vector Machine (SVM)Supervised LearningNovo Nordiskhttps://www.novonordisk.com/about/supply-chain.html, accessed on 4 January 2025
Genomic Data AnalysisConvolutional Neural Network (CNN)Supervised LearningFoundation Medicinehttps://www.foundationmedicine.com/genomic-testing/foundation-one-cdx, accessed on 4 January 2025
AI-Driven DiagnosticsRandom ForestSupervised LearningTempushttps://www.tempus.com/xai/, accessed on 4 January 2025
Drug Response PredictionGradient Boosting Machine (GBM)Supervised LearningBerg Healthhttps://www.berghealth.com/ai-driven-drug-discovery/, accessed on 4 January 2025
Patient Care IntegrationRecurrent Neural Network (RNN)Sequence PredictionPhilipshttps://www.philips.com/a-w/healthcare/solutions/healthsuite-insights, accessed on 4 January 2025
Table 5. Various applications of individual ML models.
Table 5. Various applications of individual ML models.
Machine Learning ModelApplication in Precision MedicineExampleReference
Support Vector Machines (SVMs)Classifying patients based on genetic data or identifying biomarkers associated with diseasesIdentifying genetic variants associated with breast cancer risk[5,12,113]
Random ForestsClassifying patients based on clinical data or identifying patient clustersIdentifying patient clusters based on gene expression data in lung cancer[5,12,113]
Convolutional Neural Networks (CNNs)Analyzing medical images or identifying genetic variants associated with diseasesAnalyzing brain images to identify biomarkers associated with Alzheimer’s disease[5,12,113]
Generative Adversarial Networks (GANs)Generating synthetic data or improving the quality of medical imagesGenerating synthetic CT images to improve the accuracy of liver segmentation[5,12,113]
FINDERPredicting the risk of developing a disease based on genetic and environmental factorsPredicting disease risk based on genetic and environmental factors[5,12,113]
Recurrent Neural Networks (RNNs)Analyzing sequential patient data for disease progression predictionPredicting disease progression in patients with chronic conditions[5,12,113]
Long Short-Term Memory (LSTM)Forecasting patient outcomes and treatment responsesPredicting treatment responses in cancer patients based on genomic data[5,12,113]
Decision TreesIdentifying key decision points in treatment planningGuiding treatment decisions for patients with rare genetic disorders[5,12,113]
Gradient Boosting MachinesOptimizing treatment plans based on patient-specific dataPersonalizing treatment strategies for patients with autoimmune diseases[5,12,113]
Deep Belief NetworksDiscovering complex patterns in multi-omics dataIdentifying novel biomarkers[5,12,113]
Table 6. Overview of innovations in AI and their disadvantages in pharmaceutical industry.
Table 6. Overview of innovations in AI and their disadvantages in pharmaceutical industry.
InnovationsDisadvantagesReferences
Predictive Maintenance
  • − Potential inaccuracies in failure predictions leading to unnecessary maintenance actions or overlooked issues. Complexity in data interpretation and algorithmic validation. Dependency on sensor data quality and reliability.
[138]
Production Process Optimization
  • − Challenges in algorithmic complexity and interpretability, hindering user adoption and trust. Difficulty in validating and comprehending complex machine learning models.
[139]
Demand Forecasting and Inventory Management
  • − Issues with data quality and integration, leading to inaccurate demand forecasts and suboptimal inventory management decisions. Over-reliance on historical data without accounting for market volatility.
[136]
Supply Chain Optimization
  • − Challenges related to data quality, interpretability, and scalability, impacting the effectiveness of AI-driven supply chain optimization. Dependency on accurate and timely data inputs from various sources.
[140]
Genomic Data Analysis for Personalized Treatment Strategies
  • − Ethical concerns related to data privacy and informed consent in genomic data analysis. Challenges in interpreting complex genomic data and identifying clinically actionable insights. Regulatory considerations regarding the use of genetic information in healthcare.
[141]
AI-driven Diagnostics and Biomarker Discovery
  • − Potential for algorithmic bias and inaccuracies in diagnostic predictions, leading to misdiagnosis or delayed treatment. Challenges in integrating AI-driven diagnostic tools into existing clinical workflows. Regulatory hurdles in validating and approving AI-based medical devices.
[142,143]
Integration of AI and ML in Patient Care
  • − Concerns regarding algorithmic transparency and interpretability, hindering clinician trust and adoption of AI-driven clinical decision support systems. Challenges in integrating AI tools into existing healthcare IT infrastructure and workflows. Regulatory requirements for validating AI algorithms in clinical settings
[144]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kandhare, P.; Kurlekar, M.; Deshpande, T.; Pawar, A. A Review on Revolutionizing Healthcare Technologies with AI and ML Applications in Pharmaceutical Sciences. Drugs Drug Candidates 2025, 4, 9. https://doi.org/10.3390/ddc4010009

AMA Style

Kandhare P, Kurlekar M, Deshpande T, Pawar A. A Review on Revolutionizing Healthcare Technologies with AI and ML Applications in Pharmaceutical Sciences. Drugs and Drug Candidates. 2025; 4(1):9. https://doi.org/10.3390/ddc4010009

Chicago/Turabian Style

Kandhare, Priyanka, Mrunal Kurlekar, Tanvi Deshpande, and Atmaram Pawar. 2025. "A Review on Revolutionizing Healthcare Technologies with AI and ML Applications in Pharmaceutical Sciences" Drugs and Drug Candidates 4, no. 1: 9. https://doi.org/10.3390/ddc4010009

APA Style

Kandhare, P., Kurlekar, M., Deshpande, T., & Pawar, A. (2025). A Review on Revolutionizing Healthcare Technologies with AI and ML Applications in Pharmaceutical Sciences. Drugs and Drug Candidates, 4(1), 9. https://doi.org/10.3390/ddc4010009

Article Metrics

Back to TopTop