View Article

Abstract

Artificial Intelligence (AI) is transforming the pharmaceutical industry by accelerating drug discovery, improving prediction accuracy, and reducing development costs. Traditional methods of drug design are time-consuming, costly, and often limited by human interpretation. AI integrates computational modeling, big data analytics, and machine learning (ML) to revolutionize each phase of drug discovery from target identification to clinical testing. This review explores the mechanisms, technologies, and tools behind AI in drug discovery and evaluates its potential to enhance pharmaceutical research. Applications include target validation, de novo drug design, virtual screening, and toxicity prediction. Case studies such as Alpha Fold and Scientia demonstrate how AI reduces RCD timelines and enhances molecular precision. Challenges like data bias, interpretability, and regulatory gaps are also discussed. The review concludes that integrating AI with experimental pharmacology and omics data will establish a new paradigm of precision drug discovery.

Keywords

Artificial Intelligence, Drug Discovery, Machine Learning, Deep Learning, Pharmacy, ADMET, Virtual Screening

Introduction

Drug discovery traditionally requires over a decade and billions of dollars to bring a single molecule to market. The process involves identifying potential therapeutic targets, screening compound libraries, optimizing hits, and conducting pre-clinical and clinical trials. However, the success rate remains extremely low, with only about 1 in 10,000 Compound reaching approval. AI has emerged as a powerful tool to address these inefficiencies. By mimicking human cognitive processes through algorithms, AI enables machines to analyze chemical structures, biological data, and clinical outcomes to derive meaningful insights faster than traditional computational methods. In pharmacy, this integration enhances prediction of pharmacokinetics, toxicity, and molecular interactions helping researchers focus on the most promising candidates. AI-driven discovery utilizes machine learning (ML), deep learning (DL), and natural language processing (NLP) to extract patterns from diverse datasets. Pharmaceutical companies and academic institutions increasingly adopt AI for drug repurposing, target discovery, and predictive modeling. According to a 2023 report, over 150 AI-based collaborations between pharma and tech companies have been established worldwide. [1] Drug discovery has always been a long, expensive, and uncertain scientific journey. Traditional research methods involve repeated laboratory experiments, large clinical trials, and years of trial-and-error before a single promising drug reaches the market. Because biological systems are extremely complex, researchers often struggle to predict how a new molecule will behave in the human body. As a result, despite decades of scientific progress, the majority of drug candidates still fail during development mainly due to poor efficacy, toxicity issues, or unexpected side effects (Ekins et al., 2019). In recent years, Artificial Intelligence (AI) has emerged as a revolutionary solution to these challenges. AI brings the ability to simulate biological processes, analyze millions of chemical compounds, and predict drug behavior much faster than conventional computational tools. Instead of relying solely on laboratory experiments, scientists can now use AI models to screen compounds virtually, identify molecular interactions, and make early decisions that save both time and resources. This shift represents a major transformation in how pharmaceutical research is conducted.

Fig No. 1: Drug Discovery

METHODOLOGY AND DATA SOURCES:

AI models in drug discovery depend heavily on large, high-quality datasets. Common databases include PubChem, ChEMBL, Drug Bank, and Protein Data Bank (PDB). Training data often consist of chemical descriptors, molecular fingerprints, gene expression data, and clinical trial outcomes. Data preprocessing, feature extraction, and dimensionality reduction ensure model efficiency. Algorithms such as Random Forests, Support Vector Machines (SVM), and Graph Neural Networks (GNNs) are used to correlate molecular structure with biological activity. For example, GNNs can represent molecules as graphs, where atoms are nodes and bonds are edges, allowing the network to predict binding affinity more accurately [2] However, the success of AI depends on the availability of curated, unbiased datasets. Data inconsistency or imbalance can lead to poor model generalization — a challenge discussed later in this review. AI-based drug discovery heavily depends on the quality and diversity of data used to train computational models. In traditional research, scientists manually collect and evaluate molecular information, which is often slow and prone to human bias. However, AI systems require large, well-structured datasets so that patterns related to drug activity, toxicity, and biological response can be learned accurately. This makes data collection and preprocessing one of the most critical steps in the entire drug discovery workflow. To support model training, researchers rely on publicly available chemical and biological repositories. Databases like PubChem and ChEMBL contain millions of annotated chemical structures along with biological activity results from experimental studies. Likewise, Drug Bank provides detailed information about approved and investigational drugs, including their molecular targets and pharmacological profiles. Structural databases such as the Protein Data Bank (PDB) offer 3D conformations of proteins, which are essential for structure-based AI models predicting how a drug molecule binds to its target (Berman et al., 2000). Before feeding this information into AI models, extensive preprocessing steps are required. Data cleaning removes duplicates, incorrect labels, and incomplete molecular entries. Feature extraction converts chemical meaningful numerical formats—such as fingerprints, descriptors, or graph representations—allowing machine learning algorithms to analyze molecular characteristics in a consistent way. Dimensionality reduction techniques are sometimes applied to eliminate irrelevant features and enhance model performance (Chen & Engkvist, 2023).

• Core AI Technologies in Drug Discovery:

Artificial Intelligence has introduced several powerful technologies that have changed how scientists design, analyze, and optimize drug candidates. Each AI technique contributes a unique capability, making theentire drug discovery pipeline faster, more accurate, and more efficient. Understanding these core technologies is essential because they form the foundation of modern pharmaceutical innovation.

• Machine Learning:

Machine learning uses statistical models to learn from data and make predictions. Supervised learning is widely applied for classification tasks (active vs inactive compounds), while unsupervised learning identifies hidden molecular clusters. Reinforcement learning (RL) is now used for de novo molecular design, where models learn optimal synthesis pathways by maximizing “reward” functions such as potency or solubility [3]. Machine learning enables computers to learn from previous experimental results and recognize complex patterns that are not easily identified through manual analysis. In drug discovery, ML models are trained using thousands of chemical structures and biological activities, allowing them to predict whether a molecule will be active or inactive, toxic or safe, or suitable for further development (Vamathevan et al., 2019). Supervised learning helps classify compounds and estimate potency, while unsupervised learning discovers hidden relationships among molecules. Reinforcement learning (RL), a newer approach, allows algorithms to “learn by experimentation.” RL-based platforms explore chemical properties by rewarding the model when it generates molecules with desirable characteristics—such as improved solubility or reduced toxicity (Popova et al., 2018). This makes RL a powerful tool for designing new molecular structures from scratch.

• Deep Learning:

Deep learning employs multi-layered neural networks capable of handling vast chemical and biological datasets. Convolutional Neural Networks (CNNs) and Recurrent Neural Networks (RNNs) excel in predicting molecular activity and ADMET properties. For instance, DeepChem and Chemception models have successfully predicted drug toxicity based purely on molecular structures [4]. keep learning goes a step further by using multi-layered neural networks capable of handling extremely large and complex datasets. These models are highly effective in predicting ADMET properties, protein–ligand interactions, and 3D molecular conformations.

Convolutional Neural Networks (CNNs) treat molecular structures like images, allowing them to detect spatial patterns in atoms and bonds. Recurrent Neural Networks (RNNs), on the other hand, analyze sequential data, making them useful for predicting reaction pathways or generating SMILES strings for new molecules (Goh et al., 2017). Deep learning has also enabled breakthroughs such as toxicity prediction and automated chemical feature extraction, reducing human bias and increasing prediction accuracy

• Natural Language Processing (NLP):

NLP automates literature mining, helping researchers extract drug-target relationships and clinical trial results from millions of scientific publications. OpenAI’s GPT-based models and Google’s BioBERT are now used in pharmaceutical text mining [5]. The pharmaceutical industry produces enormous amounts of scientific literature, research reports, patents, and clinical data. Natural Language Processing (NLP) helps scientists quickly extract information from these massive text sources. Modern NLP systems like BioBERT and GPT-based models can scan thousands of research papers to identify drug–target interactions, adverse effects, clinical outcomes, and disease mechanisms within seconds (Lee et al., 2022).

3. Applications of Artificial Intelligence In The Drug Discovery Pipeline:

AI technologies have transformed each stage of the drug discovery process—from target identification to clinical trials—by enabling rapid hypothesis generation and predictive modeling. Traditional experimental approaches require thousands of assays and manual data evaluation, whereas AI systems automate pattern recognition and decision-making using massive datasets. Below is an overview of the major areas where AI contributes to pharmaceutical innovation. Artificial Intelligence has reshaped the entire drug discovery process by bringing automation, predictive modeling, and data-driven decision-making to a field that traditionally relies on long experimental cycles. AI not only accelerates laboratory work but also helps scientists understand disease biology more deeply, identify promising drug molecules earlier, and minimize costly failures during later development stages. [6].

3.1 Target Identification and Validation:

The first stage in drug discovery involves identifying biological targets such as proteins, genes, or enzymes associated with disease mechanisms. AI assists by analyzing genomic, proteomic, and metabolomic data to reveal novel or hidden targets. Machine learning algorithms can correlate disease phenotypes One of the most challenging steps in drug discovery is selecting the correct biological target. A single mistake at this stage can lead to years of wasted effort. AI assists by analyzing huge datasets from genomics, proteomics, transcriptomics, and clinical research. Machine learning algorithms can uncover previously unknown disease-gene relationships and highlight which proteins are most likely to respond to drug intervention (Chen et al., 2023) AI models can also validate these targets by predicting whether they play a causal role in disease progression. This reduces dependence on trial-and-error experiments and helps researchers focus on the most promising biological pathway. [7].

3.2. Hit Discovery and Virtual Screening:

Traditionally, scientists screened millions of molecules through expensive laboratory assays. AI has transformed this process with virtual screening—a computational approach where ML and DL models rapidly evaluate chemical libraries and predict which molecules may bind effectively to a target protein. Deep learning models, such as Graph Neural Networks (GNNs), evaluate molecular interactions with high accuracy, drastically reducing the need for physical testing (Wallach et al., 2015). As a result, potential hit compounds can be identified within hours instead of months. [8].

3.3. De Novo Drug Design:

AI is now capable of designing brand-new molecular structures tailored to specific biological activities. Generative adversarial networks (GANs) and reinforcement learning algorithms create novel compounds by optimizing parameters such as potency, selectivity, toxicity, and drug-likeness. These AI-generated molecules often show better predicted performance than naturally occurring compounds, giving medicinal chemists a new starting point for drug development (Zhavoronkov et al., 2019). This innovation dramatically expands chemical space and enables the discovery of molecules that would be difficult to design manually. [9].

3.4. ADMET Prediction:

A major cause of drug failure during clinical trials is poor ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) properties. AI helps researchers predict ADMET behavior early in the pipeline, which saves time, resources, and ethicaDeep learning models trained on large toxicity and pharmacokinetic datasets can accurately estimate liver toxicity, metabolic pathways, and drug-drug interactions long before a molecule enters animal studies. [10].

4. Drug Repurposing:

AI plays a major role in finding new therapeutic uses for existing drugs. By analyzing clinical records, biological networks, and scientific literature, AI models can quickly identify unexpected drug-disease associations. During the COVID-19 pandemic, AI systems like those developed by Benevolent AI helped identify baricitinib as a potential antiviral therapy, demonstrating how repurposing powered by AI can yield rapid and effective results (Richardson et al., 2020). [11].

5. Clinical Trial Optimization:

Artificial Intelligence (AI) has become a transformative tool in modern clinical trials, helping researchers conduct studies faster, more accurately, and at a lower cost. Traditional clinical trials often face challenges such as slow patient recruitment, high operational expenses, large volumes of unstructured medical data, and delays in trial monitoring. AI addresses these challenges by automating complex tasks, analyzing patterns in clinical and biological data, and supporting data-driven decision-making throughout the trial lifecycle.AI-based systems streamline trial design, improve patient selection, enhance data monitoring, and support predictive modeling, ultimately increasing the efficiency and success rate of drug development. By integrating machine learning, natural language processing, and advanced analytics, AI is reshaping how clinical evidence is generated and evaluated. [12].

Fig No. 2: Clinical Tials

6.  Case Studies and Industrial Impact of Ai In Drug Discovery:

The integration of AI into pharmaceutical RCD is not only theoretical but has already resulted in real-world breakthroughs. Several biotech firms have demonstrated that AI can reduce drug discovery timelines from 5–6 years to less than 18 months, marking a paradigm shift in modern pharmacology. artificial Intelligence has already demonstrated its real-world value in the pharmaceutical industry through multiple successful case studies. These examples show that AI is not just a theoretical concept but a practical tool that can significantly reduce the time, cost, and uncertainty involved in developing new medicines [13].

6.1 Incilico medicens:

Insilico Medicine is a pioneer in using deep generative models and reinforcement learning to accelerate drug design. In 2019, its platform designed and synthesized a fibrosis inhibitor in just 46 days, compared to the traditional average of 4–6 years The company’s AI system, “Pharma.AI,” integrates multi-omics data, literature mining, and biological pathway modeling to identify novel targets. Insilico Medicine has become a global leader in AI-driven drug discovery. Their deep generative models are capable of creating new molecular structures with optimized therapeutic properties. One of their most famous achievements was the design of a novel fibrosis inhibitor in only 46 days—an extraordinary improvement compared to the traditional timeline of several years (Zhavoronkov et al., 2019). [14].

6.2 Exscientia:

Exscientia, a UK-based company, utilizes AI-driven automated design cycles for small- molecule discovery. Their AI platform combines Bayesian optimization with deep learning to generate and refine drug candidates in iterative loops. Exscientia partnered with Sumitomo Dainippon Pharma to design DSP-1181, a serotonin 5- HT1A receptor agonist for obsessive- compulsive disorder (OCD). The molecule advanced from project initiation to human trials in just 12 months—a world record for drug discovery efficiency [15].

6.3 Benevolent AI:

BenevolentAI applies natural language processing (NLP) and knowledge graphs to interpret millions of scientific papers and biomedical datasets. During the COVID-19 pandemic, its AI models rapidly identified baricitinib, an existing rheumatoid arthritis drug. This discovery was later validated in clinical trials, and baricitinib received emergency use authorization (EUA) by the U.S. FDA in 2020. BenevolentAI’s achievement demonstrated the potential of AI in drug repurposing and emergency response. During the COVID-19 pandemic, BenevolentAI demonstrated how AI can support global health emergencies. Their knowledge graph and text- mining platform analyzed thousands of biomedical documents to identify potential therapeutic candidates. The system flagged “baricitinib,” originally a rheumatoid arthritis drug, as a possible treatment for COVID-19 inflammation pathways (Richardson et al., 2020). This finding was later validated through experiments and clinical studies, ultimately receiving emergency use approval. This example shows how AI can speed up drug repurposing—an approach crucial during urgent health crises [16].

6.4 Atomwise:

Atomwise introduced AtomNet, the world’s first commercial deep learning neural network for structure-based drug design. Using CNNs, AtomNet predicts bioactivity from 3D molecular structure.

6.5 DeepMind’s AlphaFold:

The introduction of AlphaFold 2 by DeepMind (2020) revolutionized structural biology by achieving atomic-level accuracy in protein structure prediction [18]. AlphaFold predicted structures for over 200 million proteins, now freely available in the AlphaFold Protein Structure Database, aiding pharmacologists worldwide. This has accelerated structure-based drug design, improving target binding predictions and guiding medicinal chemists in rational molecule design. [17].

7. Impact on The Pharmaceutical Industry:

AI’s influence extends beyond individual case studies and is reshaping the entire pharmaceutical ecosystem:

7.1. Reduction in Research Costs and Timelines:

Reports indicate that AI has helped reduce early-stage discovery timelines by 60–70% and decreased R&D costs by nearly 40% (Deloitte, 2024). This allows companies to bring life- saving medicines to patients more quickly. [18].

7.2. Increased Collaboration Between Tech and Pharma:

More than 150 partnerships now exist between AI companies and major pharmaceutical leaders such as Pfizer, Novartis, Roche, and AstraZeneca. These collaborations combine biological expertise with computational power, accelerating innovation. [19]

8. Challenges and Ethical Considerations In AI-Driven Drug Discovery:

Despite its immense promise, AI in drug discovery faces numerous technical, ethical, and regulatory challenges. The integration of computational intelligence into biomedical science requires not only robust algorithms but also high-quality data, interpretability, and accountability. This section outlines the major hurdles that must be addressed to ensure safe and transparent AI deployment in pharmaceutical research [20]

8.1 Data Quality and Availability:

AI models are only as strong as the data they learn from. Pharmaceutical data are often heterogeneous, incomplete, or biased, posing serious obstacles for machine learning systems. Many biological and chemical datasets contain missing values or are derived from incompatible experimental setups. This can lead to overfitting, where the model performs well on training data but poorly on new compounds Moreover, a significant portion of pharmacological data remains proprietary, restricting access for academic researchers. Initiatives like Open Targets, ChEMBL, and the AlphaFold Database are working to democratize such information. [21].

8.2. Model Interpretability:

AI algorithms, especially deep learning models, are often referred to as “black boxes” because their decision-making processes are opaque. In drug discovery, where molecular predictions can affect patient safety, explainable AI (XAI) is critical [21]. Without interpretability, regulatory bodies like the FDA and EMA cannot evaluate or approve AI-assisted results for clinical use. Techniques such as SHAP (Shapley Additive Explanations) and LIME (Local Interpretable Model- Agnostic Explanations) are being introduced to make molecular predictions more transparent. However, balancing accuracy with interpretability remains a central challenge. [22].

Fig No. 3: Challenges in AI

9. Future Directions of AI in Drug Discovery:

While current AI applications have primarily focused on molecular prediction and optimization, future innovations are expected to redefine the entire drug discovery ecosystem. The next generation of AI-driven systems will focus on hybrid intelligence, automation, and precision therapeutics. [23].

9.1 AI and Quantum Computing for Ultra-Fast Drug Design:

Quantum computing represents a major leap in computational capability. Unlike classical computers, which process information sequentially, quantum systems can evaluate multiple molecular states simultaneously. When combined with AI, quantum machine learning (QML) may be able to simulate extremely complex molecular interactions that traditional computers cannot handle (Biamonte et al., 2021).

This will help in:

•  R&D Predicting drug-target binding with atomic-level precision

• Simulating protein dynamics in real time

• Accelerating retrosynthesis planning

• Reducing the trial-and-error cycle in early

Although quantum computing is still developing, its integration with AI could revolutionize molecular design.

9.2. Multi-Omics Integration for Precision Medicine:

Modern drug discovery is moving toward personalization. Diseases like cancer, diabetes, and autoimmune disorders vary greatly among individuals. To address this, future AI systems will integrate multi-omics data—genomics, proteomics, metabolomics, transcriptomics, and epigenomics—to create a complete biological profile of each patient (Kim et al., 2022).

This will allow AI to:

• Identify patient-specific biomarkers

• Predict individualized drug responses

• Design  targeted therapies with fewer side effects Support precision dosing recommendations [24].

9.3. Autonomous and Self-Driving Laboratories:

One of the most revolutionary ideas is the concept of self-driving laboratories—automated research environments where AI controls experiments, analyzes results, and designs the next steps without human

These labs will:

•   Automatically run high-throughput chemical reactions

•  Optimize reaction conditions using real-time feedback

•  Generate new hypotheses based on predictive modeling

• Significantly reduce manual laboratory work

This automation can speed up drug discovery by several folds, making experimental science more efficient and less labour-intensive.

9.4.  Human–AI Collaboration in Medicinal Chemistry:

Despite AI’s rapid progress, human expertise will remain essential. The future will emphasize collaborative intelligence, where chemists and AI models work together to design superior drug candidates. Medicinal chemists will guide AI by providing domain knowledge, while AI explores vast chemical spaces and suggests innovative structures (Brown & Ertl, 2023).

This partnership will result in:

• Smarter synthesis planning

• More creative molecular designs

• Better understanding of toxicity risks

• Faster optimization cycles

Rather than replacing scientists, AI will amplify their capabilities.

9.5. AI in Clinical Trials and Real-World Evidence (RWE):

Clinical trials are one of the most expensive and time-consuming stages in drug development. Future AI systems will play a key role in optimizing them by predicting patient outcomes, identifying ideal participants, and monitoring safety signals in real time. Additionally, AI will increasingly analyze real-world evidence (RWE) such as electronic health records, wearable device data, and population-level health trends[25].

CONCLUSION:

Artificial Intelligence has redefined the paradigm of modern drug discovery by offering data- driven insights, rapid molecular screening, and predictive accuracy far beyond human capability. By combining the strengths of machine learning, deep learning, and natural language processing, AI systems enable faster identification of drug targets, virtual screening of millions of compounds, and optimization of pharmacokinetic and toxicity profiles Real-world successes, such as Insilico Medicine’s AI-designed fibrosis inhibitor and Exscientia’s DSP-1181, highlight AI’s transformative role in reducing discovery timelines from several years to mere months. Meanwhile, AlphaFold’s structural prediction breakthroughs have provided the pharmaceutical community with an unprecedented understanding of protein folding and function, accelerating structure-based drug design worldwide However, challenges remain—particularly regarding data quality, algorithmic transparency, and ethical governance. The “black box” problem of deep models continues to hinder regulatory trust, while data privacy and model bias raise concerns about fairness and reproducibility. Future advancements will depend on global collaboration, open data sharing, and the development of explainable and interpretable AI systems.                                          

REFERENCE

  1. Goh GB, Siegel C, Vishnu A, Hodas N, Baker N. Chemception: A deep neural network with minimal chemistry knowledge matches the performance of expert- developed QSAR/QSPR models. ACS Cent Sci. 2017;3(8):852–859.
  2. Olivecrona M, Blaschke T, Engkvist O, Chen H. Molecular de-novo design through deep reinforcement learning. J Cheminform. 2017;9(1):48.
  3. Feinberg EN, Joshi E, Pande VS, Cheng AC. Improvement in ADMET prediction using deep learning. Front Pharmacol. 2020; 11:1588.
  4. Goh GB, Siegel C, Vishnu A, Hodas N, Baker N. Chemception: A deep neural network with minimal chemistry knowledge matches the performance of expert- developed QSAR/QSPR models. ACS Cent Sci. 2017;3(8):852–859.
  5. Olivecrona M, Blaschke T, Engkvist O, Chen H. Molecular de-novo design through deep reinforcement learning. J Cheminform. 2017;9(1):48.
  6. Feinberg EN, Joshi E, Pande VS, Cheng AC. Improvement in ADMET prediction using deep learning. Front Pharmacol. 2020; 11:1588.
  7. Wallach I, Dzamba M, Heifets A. AtomNet: A deep convolutional neural network for bioactivity prediction in structure-based drug discovery. arXiv preprint. 2015.
  8. Stokes JM, et al. A deep learning approach to antibiotic discovery. Cell. 2020;180(4):688– 702.
  9. Zhavoronkov A. Artificial intelligence for drug discovery, biomarker development, and generation of novel chemistry. Chem Soc Rev. 2018;47(2):326–341.
  10. Popova M, Isayev O, Tropsha A. Deep reinforcement learning for de novo drug design.
  11. Insilico Medicine. AI-designed preclinical candidate INS018_055 enters Phase 1 trials. Nat Biotechnol. 2021.
  12. Zhavoronkov A, et al. Deep learning for target identification and drug discovery. Mol Pharm. 2020;17(10):4146–4161.
  13. Richardson P, et al. Baricitinib as potential treatment for COVID-19: BenevolentAI discovery. Lancet. 2020;395(10223).
  14. Toney GM, et al. Explainable AI in pharmacology. Trends Pharmacol Sci. 2023;44(1):32– 46.
  15. European Commission. Ethics guidelines for trustworthy AI. EU Publications Office. 2023.
  16. U.S. FDA. Artificial Intelligence/Machine Learning-Based Software as a Medical Device Action Plan. FDA.gov. 2023.
  17. Chen H, Engkvist O. Challenges in AI-driven pharmaceutical research. Drug Discov Today. 2023;28(6):103539.
  18. Biamonte J, et al. Quantum machine learning in drug discovery. Nature. 2021;593(7858):53– 64.
  19. Biamonte J, et      al. Quantum machine learning in drug       discovery.        Nature. 2021;593(7858):53– 6.
  20. Kim S, et al. Multi-omics data integration in AI-driven pharmacology. Bioinformatics.
  21. Kim S, et al. Multi-omics data integration in AI-driven pharmacology. Bioinformatics. 2022;38(10):2673–2684.
  22. Böhm S, et al. Self-driving laboratories for drug discovery. Nat Rev Chem. 2023;7(4):245– 258.
  23. Brown N, Ertl P. Human–AI collaboration in medicinal chemistry. J Med Chem. 2023;66(14):9304–9320.
  24. Johnson KB, et al. AI-enabled precision medicine. Nat Med. 2023;29(2):271–283.
  25. Thiel WH, et al. AI in clinical trial optimization. Clin Pharmacol Ther. 2024;115(3):567– 579.

Reference

  1. Goh GB, Siegel C, Vishnu A, Hodas N, Baker N. Chemception: A deep neural network with minimal chemistry knowledge matches the performance of expert- developed QSAR/QSPR models. ACS Cent Sci. 2017;3(8):852–859.
  2. Olivecrona M, Blaschke T, Engkvist O, Chen H. Molecular de-novo design through deep reinforcement learning. J Cheminform. 2017;9(1):48.
  3. Feinberg EN, Joshi E, Pande VS, Cheng AC. Improvement in ADMET prediction using deep learning. Front Pharmacol. 2020; 11:1588.
  4. Goh GB, Siegel C, Vishnu A, Hodas N, Baker N. Chemception: A deep neural network with minimal chemistry knowledge matches the performance of expert- developed QSAR/QSPR models. ACS Cent Sci. 2017;3(8):852–859.
  5. Olivecrona M, Blaschke T, Engkvist O, Chen H. Molecular de-novo design through deep reinforcement learning. J Cheminform. 2017;9(1):48.
  6. Feinberg EN, Joshi E, Pande VS, Cheng AC. Improvement in ADMET prediction using deep learning. Front Pharmacol. 2020; 11:1588.
  7. Wallach I, Dzamba M, Heifets A. AtomNet: A deep convolutional neural network for bioactivity prediction in structure-based drug discovery. arXiv preprint. 2015.
  8. Stokes JM, et al. A deep learning approach to antibiotic discovery. Cell. 2020;180(4):688– 702.
  9. Zhavoronkov A. Artificial intelligence for drug discovery, biomarker development, and generation of novel chemistry. Chem Soc Rev. 2018;47(2):326–341.
  10. Popova M, Isayev O, Tropsha A. Deep reinforcement learning for de novo drug design.
  11. Insilico Medicine. AI-designed preclinical candidate INS018_055 enters Phase 1 trials. Nat Biotechnol. 2021.
  12. Zhavoronkov A, et al. Deep learning for target identification and drug discovery. Mol Pharm. 2020;17(10):4146–4161.
  13. Richardson P, et al. Baricitinib as potential treatment for COVID-19: BenevolentAI discovery. Lancet. 2020;395(10223).
  14. Toney GM, et al. Explainable AI in pharmacology. Trends Pharmacol Sci. 2023;44(1):32– 46.
  15. European Commission. Ethics guidelines for trustworthy AI. EU Publications Office. 2023.
  16. U.S. FDA. Artificial Intelligence/Machine Learning-Based Software as a Medical Device Action Plan. FDA.gov. 2023.
  17. Chen H, Engkvist O. Challenges in AI-driven pharmaceutical research. Drug Discov Today. 2023;28(6):103539.
  18. Biamonte J, et al. Quantum machine learning in drug discovery. Nature. 2021;593(7858):53– 64.
  19. Biamonte J, et      al. Quantum machine learning in drug       discovery.        Nature. 2021;593(7858):53– 6.
  20. Kim S, et al. Multi-omics data integration in AI-driven pharmacology. Bioinformatics.
  21. Kim S, et al. Multi-omics data integration in AI-driven pharmacology. Bioinformatics. 2022;38(10):2673–2684.
  22. Böhm S, et al. Self-driving laboratories for drug discovery. Nat Rev Chem. 2023;7(4):245– 258.
  23. Brown N, Ertl P. Human–AI collaboration in medicinal chemistry. J Med Chem. 2023;66(14):9304–9320.
  24. Johnson KB, et al. AI-enabled precision medicine. Nat Med. 2023;29(2):271–283.
  25. Thiel WH, et al. AI in clinical trial optimization. Clin Pharmacol Ther. 2024;115(3):567– 579.

Photo
Dr. Sunil Jaybhayee
Corresponding author

Faculty of Pharmacy, Dr. Babasaheb Ambedkar Technological University, Raigad, Lonere

Photo
Payal Kundkar
Co-author

Faculty of Pharmacy, Dr. Babasaheb Ambedkar Technological University, Raigad, Lonere

Photo
Nikita Pungle
Co-author

Faculty of Pharmacy, Dr. Babasaheb Ambedkar Technological University, Raigad, Lonere

Dr. Sunil Jaybhaye*, Payal Kundkar, Nikita Pungle, Artificial Intelligence in Drug Discovery: Challenges and Future Direction in Pharmaceutical Research, Int. J. Sci. R. Tech., 2025, 2 (12), 224-232. https://doi.org/10.5281/zenodo.17929314

More related articles
Formulation and Evaluation of Polyherbal Antidiabe...
Tushar Awhad , Pritam Khurde , Dr. Jayshri Bairagi, ...
Smart Polymers is Controlled Drug Release...
Shivaji Katade , Nayan Anil Sardar, Ram Baban Ingle, ...
A Review on the Role of Transdermal Drug Delivery:...
Yash Tambe, Vaishali Pagar, Harshal Pagar, Vaibhav Thakare, ...
Emerging Multidrug-Resistant Fungal Pathogens: Epidemiology, Mechanisms, and Nov...
Haider Abbas, Amulya Singh, Arpit Maurya , Kavya Singh, Dr. Anupam Singh, ...
Characterizing the Role of Suzetrigine Drug as A Non-Opioid Analgesic for Acute ...
Shraddha Ghadage, Snehal Kadbhane, Dr. Vijaykumar kale, Shweta Mahajan, Shweta Dandawthe, ...
An Artificial Intelligence in Pharmaceutical Sciences: Current Trends, Applicati...
Ruswa Urade, Sujata Samant, Sandip Umare, Rupal Kalbhut, Bhudevi Khapne, ...
Related Articles
Artificial Intelligence in Clinical Trials...
Utkarsha Mane, Dr. Mahesh Thakare, Rushikesh Telore, Vaishnavi Hendge, Bhagyashri Thorbole, ...
Reinventing Medicines: Drug Repurposing as A New Frontier in Cancer Therapy...
Tejswini Gaikwad, Rashee Shahu, Sunita Kode, Shivcharan Kamble, Pooja Rasal, ...
A Review Article On: Gastro-Retentive Drug Delivery System (GRDDS)...
Achal Andhale, Sakshi Kadam, Mansi Jamdhade, Rutuja Jamdhade, Jyoti Javade, ...
More related articles
Smart Polymers is Controlled Drug Release...
Shivaji Katade , Nayan Anil Sardar, Ram Baban Ingle, ...
A Review on the Role of Transdermal Drug Delivery: Microneedles, Patches, and Na...
Yash Tambe, Vaishali Pagar, Harshal Pagar, Vaibhav Thakare, ...
Smart Polymers is Controlled Drug Release...
Shivaji Katade , Nayan Anil Sardar, Ram Baban Ingle, ...
A Review on the Role of Transdermal Drug Delivery: Microneedles, Patches, and Na...
Yash Tambe, Vaishali Pagar, Harshal Pagar, Vaibhav Thakare, ...