v
Search
Advanced

Publications > Journals > Journal of Exploratory Research in Pharmacology> Article Full Text

  • OPEN ACCESS

Artificial Intelligence-driven Strategies and Modern Innovations to Surpass Biopharmaceutic Limitations in Traditional Drug Development

  • Haneen Badreldin Ali1,*  and
  • Muhammad Burhan Khan2 
 Author information 

Abstract

Drug discovery is an exceptionally long and costly process, often taking over 10 years and costing billions of dollars. Despite these efforts, more than 90% of drug candidates fail, with most failures occurring during clinical trials due to issues related to efficacy, safety, or poor pharmacokinetics. A major contributor to these failures is biopharmaceutic barriers, including poor solubility, limited permeability, active efflux by transporters such as P-glycoprotein and breast cancer resistance protein, and extensive first-pass metabolism by CYP450 enzymes. These factors severely limit drug absorption and bioavailability, reducing therapeutic efficacy. Although traditional approaches, such as high-throughput absorption, distribution, metabolism, and excretion screening and improved chemical design, have achieved some progress, a major shift is now occurring through the use of in silico modeling, artificial intelligence (AI), and machine learning. These AI-driven tools enhance the prediction accuracy of absorption, distribution, metabolism, and excretion profiles, identify transporter interactions, and even simulate metabolic pathways. Additionally, modern formulation technologies, such as three-dimensional printing, lipid-based nanocarriers, and biodegradable delivery systems, are increasingly being integrated with AI-powered design platforms to personalize and optimize drug delivery. However, these promising advancements also raise regulatory and ethical concerns that must be addressed before widespread adoption. This review examines the major biopharmaceutic barriers responsible for drug development failures and explores how emerging AI-driven strategies and formulation innovations are being used to overcome these limitations. It also discusses current regulatory challenges and ethical considerations associated with adopting these technologies.

Keywords

Biopharmaceutics, Drug development, Artificial intelligence in drug design, In silico ADME, 3D-printed dosage forms, Biodegradable nanocarriers

Introduction

The path to discovering and developing a new drug is far from easy. It is a process that can take over a decade and cost upwards of 1 to 2 billion dollars,1 yet the success rate remains shockingly low. More than 90% of drug candidates fail during clinical development, and if preclinical failures are included, the odds are even worse.2 While many of these failures are attributed to lack of efficacy or safety issues such as toxicity, a substantial proportion are actually due to poor biopharmaceutic properties. In other words, a drug may appear promising in theory, but if it does not dissolve well, cannot cross biological membranes, is actively pumped out by efflux transporters, or is metabolized too quickly, it will not perform effectively in the body. These challenges fall under the domain of biopharmaceutics, the study of how a drug’s formulation and physicochemical properties affect its absorption and bioavailability. Despite ongoing advances in medicinal chemistry and formulation science, many compounds with therapeutic potential fail due to unresolved issues related to solubility, permeability, transport, or metabolism.3,4 The need for smarter, predictive strategies grows as drug molecules become more complex. Emerging technologies such as in silico modeling, machine learning (ML), and artificial intelligence (AI) now offer powerful tools to assess biopharmaceutical behavior early in development. Innovations in drug delivery systems, including lipid-based carriers, nanotechnology, and three-dimensional printing (3DP), have also reshaped the formulation of poorly bioavailable drugs. AI is playing a growing role in drug development, particularly in handling complex early-stage decisions. ML, one of the most widely used AI approaches, involves models that learn from data to make predictions about absorption, distribution, metabolism, and excretion (ADME), guide structural optimization, or classify drug candidates.5,6 An advanced form of ML called deep learning can recognize patterns in large and complex datasets, detecting genetic information and molecular structures that would be difficult to identify manually. Natural language processing is also being used to analyze massive amounts of biomedical literature and clinical trial data to uncover connections that might otherwise be missed. Additionally, experts and generative models are being employed to design drugs and predict molecular behavior. These AI techniques are making pharmaceutical research faster, more accurate, and more efficient, as illustrated in Figure 1. The goal of this review is to provide a comprehensive overview of the major biopharmaceutic barriers to drug success, including poor solubility, limited permeability, transporter-mediated efflux, and extensive metabolism. It also discusses recent technological developments and computational innovations aimed at overcoming these challenges, as well as regulatory considerations and future directions for integrating these approaches into modern drug development.

The role of AI in biopharmaceuticals utilizes in silico modeling, machine learning, and deep learning to enhance ADME properties and improve drug bioavailability and development success.
Fig. 1  The role of AI in biopharmaceuticals utilizes in silico modeling, machine learning, and deep learning to enhance ADME properties and improve drug bioavailability and development success.

ADME, absorption, distribution, metabolism, and excretion; AI, artificial intelligence; SVM, support vector machine.

Biopharmaceutic barriers to drug success

Drug discovery and development is an extremely long and expensive process, taking up to 10 years and costing over one to two billion dollars. Despite these investments, more than 90% of drug candidates fail during clinical development. What makes this even more concerning is that this 90% failure rate only applies to drugs that actually make it into clinical trials. If preclinical failures were included, the odds of success would be even lower.7,8 The main reasons behind these failures include lack of efficacy, which accounts for approximately 40–50% of cases, followed by safety-related issues such as toxicity (around 30%). In addition, poor drug-like properties, including low solubility, limited permeability, or extensive metabolism, contribute to about 10–15% of failures. Although significant progress has been made in optimizing drug-like properties, the overall clinical success rate has not improved considerably and still remains low, at around 10–15%.7

Solubility and permeability issues

Although a drug can be pharmacologically effective in vitro, it must first dissolve in gastrointestinal fluids and traverse the intestinal epithelium to exert therapeutic effects when administered orally. Solubility is the first and most critical requirement for absorption, and its absence often results in limited or erratic bioavailability. Drugs must cross the gut wall to reach the bloodstream, which becomes difficult if they are too hydrophilic or too large.9 In fact, it is estimated that approximately 40% of approved drugs and nearly 90% of drug candidates in development are poorly water-soluble, a condition that severely affects their clinical success and market translation.10–12 The Biopharmaceutics Classification System (BCS) was developed to conceptualize the relationship between solubility and permeability. According to this system, compounds are classified into four classes based on their solubility in water and their permeability across the gut wall.13,14 For example, Class I medications, such as propranolol, are highly soluble and highly permeable, and therefore typically do not encounter significant absorption problems. Class II drugs, like carbamazepine, have good permeability but poor solubility, so formulation strategies such as solid dispersions, amorphous forms, or lipid-based delivery systems must be employed.15 In contrast, BCS Class III drugs, such as alendronate and acyclovir, dissolve readily but are poorly absorbed due to limited membrane permeability, necessitating bioenhancers or advanced delivery systems such as microparticles and liposomes.16 BCS Class IV compounds, which lack both solubility and permeability, pose substantial formulation challenges and often exclude oral administration unless innovative technologies are used.15 To overcome absorption limitations in these classes, lipid-based nanocarriers, solid dispersions, and 3D-printed microstructures are actively being investigated. Computational tools, including quantitative structure-activity relationship models, ML, and AI, now allow molecular libraries to be screened for solubility and permeability characteristics at high throughput, streamlining formulation design and candidate selection.17 This integrated approach reduces development costs and late-stage attrition caused by unfavorable biopharmaceutical properties. Unlike traditional solubility screening methods, which are labor-intensive and require empirical formulation development, AI-based tools such as ADMETlab 2.0 and SwissADME can predict solubility profiles, BCS classes, and permeability in real time with high accuracy.18,19 Compared with standard in vitro screens, these platforms provide better insights for formulation decisions at an earlier stage of compound triaging, as illustrated in Figure 2.

Comparison of traditional and AI-based drug development strategies shows that AI approaches utilize predictive models, while traditional methods depend on iterative laboratory testing.
Fig. 2  Comparison of traditional and AI-based drug development strategies shows that AI approaches utilize predictive models, while traditional methods depend on iterative laboratory testing.

ADME, absorption, distribution, metabolism, and excretion; AI, artificial intelligence.

Transporters like P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP)

Even if a drug dissolves efficiently and demonstrates good permeability, it still faces another obstacle: efflux transporters. Proteins such as P-gp and BCRP are present in the intestinal lining and can significantly affect drug absorption. These transporters, often referred to as “cellular bouncers”,20 actively export drugs back into the intestinal lumen, thereby reducing both absorption and overall bioavailability.21 Drugs such as digoxin, paclitaxel, and topotecan are classic examples affected by P-gp and BCRP.22–24 These transporters not only influence intestinal absorption but also play crucial roles in drug distribution and elimination. P-gp and BCRP are expressed in multiple tissues, including the liver, kidney, placenta, and blood-brain barrier. Their physiological function is to protect tissues from xenobiotics; however, this protective mechanism can severely limit the accessibility of therapeutic agents to target sites. Additionally, genetic polymorphisms in transporter genes such as ABCB1 and ABCG2 can alter transporter expression, affecting the pharmacokinetics and therapeutic response of substrates.25,26 In cancer therapy, overexpression of P-gp in tumor cells can lead to multidrug resistance, where cancer cells become resistant to chemotherapy despite high drug doses.27 To study efflux-mediated transport, the Caco-2 cell line is widely used due to its similarity to human enterocytes. However, differentiation and formation of tight junctions take 21–24 days, making it less suitable for high-throughput screening.28 AI-based tools such as quantitative structure–property relationship modeling have emerged as efficient alternatives, capable of predicting permeability and efflux interactions in silico. Platforms like DeepTox and other ML-based transporter prediction models can screen large compound libraries for P-gp and BCRP affinity, enabling early detection of liabilities and reducing the need for extensive in vitro testing.29 In addition to P-gp and BCRP, organic cation transporters and organic anion transporters (OATs) play important clinical roles. They are primarily expressed in the kidney and liver, where they regulate systemic availability and clearance of many therapeutics.21,30 For example, organic cation transporter 2 mediates the renal uptake of metformin31 and cisplatin32; its inhibition or genetic variation can alter drug efficacy and toxicity.33 Similarly, OAT1 and OAT3 facilitate the uptake and elimination of penicillin, methotrexate, and NSAIDs, making them critical determinants of drug half-life and renal clearance.34 Understanding whether a drug is a substrate or inhibitor of these transporters is now considered a critical component of preclinical evaluation. Regulatory agencies, including the U.S. Food and Drug Administration (FDA) and the European Medicines Agency, increasingly require comprehensive in vitro and in vivo data on transporter interactions.35 This is essential for predicting and managing drug-drug interactions, which may result in enhanced toxicity or therapeutic failure. Advanced in silico models and AI-based prediction platforms are also used to detect transporter-related liabilities early in development.36,37 By identifying transporter interactions early and integrating this data with pharmacokinetic modeling, developers can optimize dosing strategies, anticipate population variability, and improve the safety and efficacy profile of novel therapeutics.38

Metabolism and CYP450 enzymes

Another critical barrier to drug bioavailability is metabolism. Even if a drug is absorbed, it can be extensively metabolized by liver enzymes before reaching systemic circulation, diminishing its therapeutic efficacy. This phenomenon, known as first-pass metabolism, is primarily carried out by the cytochrome P450 family, especially CYP3A4, CYP2D6, and CYP2C9, which together metabolize approximately 80% of commonly used drugs.39 A classic example is propranolol, which has only about 25% bioavailability due to significant hepatic metabolism.40 Similarly, midazolam is almost entirely metabolized by CYP3A4.41 One of the most significant challenges posed by CYP-mediated metabolism is its high interindividual variability, largely due to genetic polymorphisms. These variations can drastically influence drug exposure and patient response, classifying individuals as “poor metabolizers” or “ultra-rapid metabolizers,” which increases the risk of side effects or therapeutic failure.42 Co-administered drugs or foods can also inhibit or induce CYP enzymes, leading to dangerous drug-drug interactions. A notable example is grapefruit juice, which contains furanocoumarins that inhibit intestinal CYP3A4, resulting in elevated plasma levels of simvastatin and potentially causing myopathy and hepatotoxicity.43 Modern drug development increasingly integrates computational approaches to address these challenges. AI-enabled tools such as SMARTCyp, DeepCYP, and MetaSite utilize molecular characteristics and structure-activity relationships to predict vulnerable sites of metabolism, assisting medicinal chemists in designing metabolically stable drug candidates.44–46 Integrating predictive metabolism modeling during early-stage development enhances candidate selection, minimizes late-stage failures, and supports rational dose design,39 particularly in pharmacogenetically diverse populations.47

AI & computational modeling in biopharmaceutics

ADME & in silico models

The optimization of drug therapy depends on the precise prediction of how a medication is ADME within the body. Biopharmaceutics integrates knowledge from pharmacology, chemistry, and physiology to optimize drug discovery and development. At its core, biopharmaceutics studies how a drug’s formulation influences its ADME, ultimately impacting efficacy and safety.48,49 Unfavorable ADME properties are a major cause of late-stage drug candidate failure, resulting in significant financial losses and time expenditure in research and development.17 To address this challenge, in silico modeling was introduced, with its earliest models dating back to the 1970s–1980s. These models primarily employed quantitative structure-activity relationships to predict drug-receptor interactions based on molecular properties.50 In silico models use computational techniques to develop pharmacological models that predict biological and chemical processes, thereby aiding drug discovery and development. Increasing computational power has expanded the scope of in silico modeling, reducing costs and improving efficiency in the drug discovery process.17,51

ADME screening methods

ADME screening is used to identify and optimize new chemical entities, the primary focus of drug discovery. Traditional ADME screening methods relied on low-throughput in vitro and in vivo assays, which were manual and inefficient.52 Around the year 2000, high-throughput in vitro ADME (HT-ADME) methods were introduced. While still relying on biological assays, HT-ADME operates on an industrial scale using robotics, advanced analytical techniques such as LC/MS/MS, liquid handlers, and miniaturized assay plates, making the process more efficient and cost-effective than low-throughput methods. However, HT-ADME is still limited compared with in silico models, as it relies on physical experiments, expensive equipment, consumables, and considerable time to generate data.53,54

Advantages of AI models in ADME prediction

AI models in in silico screening provide significant advantages, particularly in predictive accuracy and efficiency. Using computational methods such as ML, these models can analyze large datasets to identify complex patterns related to pharmacokinetic properties. By simplifying relationships between physicochemical parameters and ADME endpoints, AI enables rapid identification of promising candidates without extensive wet lab experiments.51 These models enhance new chemical entity design by simultaneously optimizing multiple ADME parameters alongside bioactivity.55,56

One such model is SwissADME, a free web tool with a user-friendly interface that provides access to robust predictive models for pharmacokinetics, physicochemical properties, drug-likeness, and medicinal chemistry friendliness. SwissADME stands out for its advanced prediction algorithms, including the BOILED-Egg model, bioavailability radar, and Lipinski’s Rule of Five. These algorithms serve diverse purposes, from evaluating drug-likeness to predicting gastrointestinal absorption and blood-brain barrier permeability.18,57

Another recent model, Deep-Pk, uses graph neural networks and graph-based signatures to deliver strong predictive performance across multiple endpoints. Deep-Pk predicts ADMET (absorption, distribution, metabolism, excretion, and toxicity) across 73 endpoints, achieving an external validation mean Matthews correlation coefficient of 0.58 and receiver operating characteristic – area under the curve (ROC-AUC) scores exceeding 0.80 for most classification tasks.58 However, like many deep learning models, Deep-Pk may struggle with interpretability and could exhibit bias when applied to chemical spaces differing from its training set. kMoL, an open-source ML library, has demonstrated strong performance on benchmark datasets, achieving ROC-AUC scores above 0.75 in ADME and toxicity tasks, both in centralized and federated settings.59 By using graph convolutional networks to model molecular structures as graphs, kMoL outperforms traditional methods.59,60 Despite its flexibility and focus on privacy preservation, performance can vary depending on dataset size and quality, making careful hyperparameter tuning essential. While uncertainties remain in in silico predictions, particularly in drug discovery, methods to quantify these uncertainties can enhance interpretability and improve confidence in model predictions.61

ML applications in formulation design

ML, a branch of AI, creates predictive models trained on previous datasets, which can be derived from both in vivo and in vitro experiments. ML-based approaches are increasingly common, providing significant predictive power and screening capabilities that help identify the most suitable drug candidates, thereby enhancing accuracy and efficiency.62 The most commonly used ML techniques for screening are support vector machines (SVMs) and random forests (RFs). SVMs excel in high-dimensional spaces by identifying hyperplanes that maximize class separation, but they require careful tuning and are less efficient with large datasets. In contrast, RFs use multiple decision trees to handle large or noisy datasets with minimal tuning, offering resistance to overfitting. Although RFs may lack some interpretability compared to SVMs, they are better suited for diverse real-world data. SVMs work best with structured data, while RFs are preferred for scalability.63,64

Both techniques are widely employed in drug formulation design, particularly for classification and regression tasks. SVMs are highly effective for binary classification and high-dimensional datasets, making them suitable for predicting ADME properties such as solubility. ML-based models also analyze diverse genomic, proteomic, and clinical datasets to identify potential drug targets and prioritize candidates based on criteria such as drug-likeness and therapeutic relevance.62,65 The AlphaFold program, developed by DeepMind, has transformed AI-driven drug discovery by predicting 3D protein structures with unmatched accuracy through deep learning. AlphaFold aids structure-based drug design by identifying protein-ligand interaction sites, supporting target validation and virtual screening. With over 200 million structures available in the AlphaFold Protein Structure Database, it has become an essential resource in rational drug design.66,67 As target structure prediction improves, efficient screening of vast compound libraries becomes increasingly important in identifying optimal candidates.

Modern drug discovery heavily relies on compound libraries containing millions of molecules. For example, the ZINC22 library houses 4.1 billion molecules. The growing size of these libraries necessitates faster and more efficient screening methods.68 Early ML models using RFs focused on computational prioritization but lacked scalability.69 The sheer volume of data from genomics, proteomics, metabolomics, imaging, and electronic health records requires AI to discern meaningful patterns, process large datasets efficiently, and analyze longitudinal patient data. This capability enables understanding of individual patient trajectories, which is essential for predicting how a drug is absorbed and metabolized over time.70

How does AI predict and personalize drug absorption?

AI has become increasingly important in screening compounds with favorable ADME properties. One recent advancement is RosettaVS, an AI-driven platform that uses physics-based protocols to dock billion-molecule libraries in days, achieving a 44% hit rate for Nav1.7 in just seven days.71 AI and ML also play a crucial role in understanding the complex relationship between an individual’s genetic makeup and drug response. In pharmacogenomics, genetic variations strongly influence drug metabolism and transport, directly affecting absorption. AI can analyze an individual’s genetic profile to identify genes responsible for drug absorption and metabolism, predict likely drug responses, and enable selection of the safest and most effective treatment. AI-powered pharmacogenomics has the potential to revolutionize personalized medicine, providing innovative solutions and improved predictive accuracy to benefit patient care.72,73 However, AI tools must undergo thorough evaluation to ensure they are applied safely and fairly, just as with any new therapeutic intervention.74

Biopharmaceutics

3D-printed pharmaceuticals for personalized bioavailability

Drug discovery has seen remarkable progress, yet scientific innovation continues to emerge rapidly across multiple fields. One such innovation is 3DP, a process that creates a three-dimensional object from a digital design. Unlike traditional subtractive manufacturing, which removes material, 3DP builds objects layer by layer,75 3DP enables customization of drug delivery systems, providing personalized treatment options. It allows the creation of complex dosage forms, such as multi-compartmental capsules, which can combine multiple drugs in a single dosage form and be adjusted according to combination therapy needs.76,77 While 3DP shows great potential for improving drug bioavailability and facilitating personalized therapies, most current data comes from in vitro experiments or animal studies. Large-scale clinical trials validating these methods are limited, and regulatory approval pathways are still evolving.78,79 3DP also offers advantages for formulating drugs with poor water solubility.80 Mesoporous materials can enhance the dissolution rate and bioavailability of such drugs using techniques such as fused deposition modeling or semi-solid extrusion (SSE). For example, drug-loaded mesoporous magnesium carbonate formulations have achieved high drug loading of 15.3% w/w.81

Techniques such as fused deposition modeling, stereolithography, selective laser sintering, binder jetting, SSE, and hot-melt extrusion offer diverse possibilities in pharmaceuticals.82 For instance, SSE can produce tablets and suppositories with enhanced dissolution kinetics, increasing bioavailability by extruding a non-aqueous paste containing drug-loaded mesoporous materials.83 Despite its potential, 3DP faces limitations, including material selection, biocompatibility concerns, potential toxicity from degradation products such as those from polylactic acid, regulatory hurdles, scalability issues, and cost-effectiveness. Customization of dosages also raises ethical concerns, as it relies on sensitive patient data that must be protected.84 Nevertheless, the future of 3DP remains promising, especially when integrated with AI and ML to further expand its applications.85

Biodegradable nanocarriers for targeted drug delivery

Nanocarriers are another promising avenue in drug discovery. These nanoscale delivery systems encapsulate and transport therapeutic agents, fine-tuning drug ADME profiles.86 Biodegradable nanomaterials used in nanocarrier technology hold particular promise, as they safely degrade in the body into natural elements over time, reducing systemic toxicity. Nanocarriers are typically composed of polymers, both natural and synthetic. Natural polymers include polysaccharides and proteins such as chitosan, alginates, albumin, and cellulose derivatives. Synthetic polymers, such as polylactic acid, are popular for their high biodegradability.87 Nanocarriers provide targeted drug delivery through passive and active mechanisms. Passive targeting relies on inherent nanocarrier properties to accumulate in specific areas, such as tumor sites. Active targeting involves techniques such as ligand-mediated targeting or molecularly imprinted nanocarriers, facilitating site-specific drug delivery.88 Despite these advances, concerns remain regarding long-term biocompatibility, potential toxicity from accumulation or immune responses, and challenges in large-scale manufacturing, limiting clinical translation.89,90 For example, poly(lactic-co-glycolic acid)–polyethylene glycol (PLGA-PEG) nanoparticles co-encapsulating gemcitabine and botulinic acid have demonstrated improved anti-tumor efficacy in pancreatic cancer cell lines.91

Regulatory challenges in biopharmaceutical innovation

Although addressing biopharmaceutical needs by employing AI and other technologies shows promise, significant regulatory challenges remain. AI and ML heavily depend on representative data, but pharmaceutical datasets are complex and often uncertain, which can introduce bias in AI training and lead to inaccurate outcomes, potentially exacerbating health disparities. Ensuring data quality, reproducibility, and reliability is therefore critical.92 Regulatory agencies, such as the FDA and the European Medicines Agency, are developing frameworks to govern AI use, incorporating real-world and open-source data along with robust quality standards to ensure fair and ethical implementation in drug discovery.93 Innovations such as 3DP and biodegradable nanocarriers also face lengthy approval processes. Preclinical and clinical data are required to demonstrate safety and efficacy, imposing significant cost burdens. For 3DP, consistent quality, scalable production, and material safety, including inks used in printing, must be ensured. For nanocarriers, challenges include characterizing nanomaterials’ size, shape, and surface properties, minimizing off-target accumulation, and ensuring biocompatibility and safety.

Implementation strategies for overcoming regulatory challenges in novel drug formulations

To overcome these challenges, active communication with regulatory authorities is essential during development. The FDA’s Emerging Technology Program, for example, provides developers with real-time guidance on novel manufacturing practices, aligning preclinical and clinical protocols with regulatory expectations and reducing issues in later stages.94 Adopting quality by design principles allows manufacturers to build quality into the product from the outset, simplifying critical quality attributes and critical process parameters to ensure consistency and reproducibility.95 Regulatory agencies, following ICH Q8–Q11 guidelines, endorse quality by design principles to streamline approvals for innovative products. By adopting these strategies, cutting-edge therapies can reach patients without compromising safety or efficacy.96,97

Limitations and future prospects

The incorporation of AI, advanced predictive tools, and innovative drug formulation methods can help address the complex challenges associated with drug development. Future research should focus on validating AI-based prediction tools for drug ADME using extensive real-world patient datasets, enhancing regulatory acceptance of these algorithms.98,99 Additionally, integrating AI with 3DP and designing nanocarriers creates new opportunities for personalized medicine, enabling drug delivery tailored to individual patient profiles. At the same time, it is essential to use data ethically, minimize biases in AI programs, and ensure transparency in AI-driven decisions throughout the drug development process.

Several limitations of this review should also be acknowledged. Given the rapid evolution of this field, some of the latest developments may not be fully covered, and the review aims to provide an overview of key biopharmaceutical barriers and emerging AI-driven strategies. While case studies and tool-specific examples are presented, many applications remain in preclinical or experimental stages and have not yet been tested in large populations. Furthermore, this review provides limited coverage of health equity implications, global regulatory harmonization, and the economic feasibility of adopting AI and advanced formulation technologies. These areas warrant further investigation in the near future.

Conclusions

Biopharmaceutic challenges, including poor solubility, limited permeability, active drug efflux, and extensive metabolism, remain major reasons why many promising drug candidates fail to reach the market. While traditional formulation approaches aim to improve biopharmaceutical properties, they often overlook modern, multi-targeted, and chemically complex molecules. Addressing these obstacles in contemporary research requires integrating advanced technologies. In early stages of drug development, AI is increasingly applied to predict absorption and metabolism profiles using tools like SwissADME and DeepCYP, while generative models assist in synthesizing drug-like molecules with improved target characteristics. In silico simulations of transporter interactions also support risk evaluations prior to clinical trials. In drug formulation, 3DP enables the creation of multi-compartmental and layered tablets tailored to specific absorption windows and desired release kinetics. Meanwhile, lipid-based and PLGA-PEG nanoparticles serve as biodegradable carriers, improving the delivery of poorly soluble and unstable drugs in oncology and central nervous system therapies that require advanced delivery systems. These examples demonstrate that the application of these innovations is no longer a distant potential; they are already making meaningful impacts. As integration continues, it will be essential to align these tools with regulatory frameworks, ensure data transparency, and focus on clinically relevant endpoints to ensure their long-term impact on successful drug development.

Declarations

Acknowledgement

None.

Funding

None.

Conflict of interest

The authors have no conflict of interest related to this publication.

Authors’ contributions

Conceptualization (HBA, MBK), literature search, writing – original draft (HBA), data interpretation, supervision, critical revision of the manuscript (MBK), writing – reviewing and editing (HBA, MBK). Both authors have approved the final version and publication of the manuscript.

References

  1. Hughes JP, Rees S, Kalindjian SB, Philpott KL. Principles of early drug discovery. Br J Pharmacol 2011;162(6):1239-1249 View Article PubMed/NCBI
  2. Amorim AMB, Piochi LF, Gaspar AT, Preto AJ, Rosário-Ferreira N, Moreira IS. Advancing Drug Safety in Drug Development: Bridging Computational Predictions for Enhanced Toxicity Prediction. Chem Res Toxicol 2024;37(6):827-849 View Article PubMed/NCBI
  3. Nyamba I, Sombié CB, Yabré M, Zimé-Diawara H, Yaméogo J, Ouédraogo S, et al. Pharmaceutical approaches for enhancing solubility and oral bioavailability of poorly soluble drugs. Eur J Pharm Biopharm 2024;204:114513 View Article PubMed/NCBI
  4. Wen H, Jung H, Li X. Drug Delivery Approaches in Addressing Clinical Pharmacology-Related Issues: Opportunities and Challenges. AAPS J 2015;17(6):1327-1340 View Article PubMed/NCBI
  5. Bao Z, Bufton J, Hickman RJ, Aspuru-Guzik A, Bannigan P, Allen C. Revolutionizing drug formulation development: The increasing impact of machine learning. Adv Drug Deliv Rev 2023;202:115108 View Article PubMed/NCBI
  6. Fleming N. How artificial intelligence is changing drug discovery. Nature 2018;557(7707):S55-S57 View Article PubMed/NCBI
  7. Sun D, Gao W, Hu H, Zhou S. Why 90% of clinical drug development fails and how to improve it?. Acta Pharm Sin B 2022;12(7):3049-3062 View Article PubMed/NCBI
  8. Mordwinkin NM, Lee AS, Wu JC. Patient-specific stem cells and cardiovascular drug discovery. JAMA 2013;310(19):2039-2040 View Article PubMed/NCBI
  9. Conradi RA, Burton PS, Borchardt RT. Physico-Chemical and Biological Factors that Influence a Drug’s Cellular Permeability by Passive Diffusion. Lipophilicity in Drug Action and Toxicology. In: PliÅ¡ka V, Testa B, van de Waterbeemd H (eds). Lipophilicity in Drug Action and Toxicology. Weinheim: Wiley-VCH; 2008:233-252 View Article
  10. Xie B, Liu Y, Li X, Yang P, He W. Solubilization techniques used for poorly water-soluble drugs. Acta Pharm Sin B 2024;14(11):4683-4716 View Article PubMed/NCBI
  11. Kalepu S, Nekkanti V. Insoluble drug delivery strategies: review of recent advances and business prospects. Acta Pharm Sin B 2015;5(5):442-453 View Article PubMed/NCBI
  12. Savjani KT, Gajjar AK, Savjani JK. Drug solubility: importance and enhancement techniques. ISRN Pharm 2012;2012:195727 View Article PubMed/NCBI
  13. Yu LX, Amidon GL, Polli JE, Zhao H, Mehta MU, Conner DP, et al. Biopharmaceutics classification system: the scientific basis for biowaiver extensions. Pharm Res 2002;19(7):921-925 View Article PubMed/NCBI
  14. Amidon GL, Lennernäs H, Shah VP, Crison JR. A theoretical basis for a biopharmaceutic drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability. Pharm Res 1995;12(3):413-420 View Article PubMed/NCBI
  15. Yasir M, Asif M, Kumar A, Aggarval A. Biopharmaceutical Classification System: An Account. Int J PharmTech Res 2010;2(3):1681-1690
  16. Asad M, Rasul A, Abbas G, Shah MA, Nazir I. Self-emulsifying drug delivery systems: A versatile approach to enhance the oral delivery of BCS class III drug via hydrophobic ion pairing. PLoS One 2023;18(6):e0286668 View Article PubMed/NCBI
  17. Wu F, Zhou Y, Li L, Shen X, Chen G, Wang X, et al. Computational Approaches in Preclinical Studies on Drug Discovery and Development. Front Chem 2020;8:726 View Article PubMed/NCBI
  18. Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep 2017;7:42717 View Article PubMed/NCBI
  19. Cheng F, Li W, Zhou Y, Shen J, Wu Z, Liu G, et al. admetSAR: a comprehensive source and free tool for assessment of chemical ADMET properties. J Chem Inf Model 2012;52(11):3099-3105 View Article PubMed/NCBI
  20. Epel D, Luckenbach T, Stevenson CN, Macmanus-Spencer LA, Hamdoun A, Smital T. Efflux transporters: newly appreciated roles in protection against pollutants. Environ Sci Technol 2008;42(11):3914-3920 View Article PubMed/NCBI
  21. Giacomini KM, Huang SM, Tweedie DJ, Benet LZ, Brouwer KL, Chu X, et al. Membrane transporters in drug development. Nat Rev Drug Discov 2010;9(3):215-236 View Article PubMed/NCBI
  22. Lee J, Kang J, Kwon NY, Sivaraman A, Naik R, Jin SY, et al. Dual Inhibition of P-gp and BCRP Improves Oral Topotecan Bioavailability in Rodents. Pharmaceutics 2021;13(4):559 View Article PubMed/NCBI
  23. Husain A, Makadia V, Valicherla GR, Riyazuddin M, Gayen JR. Approaches to minimize the effects of P-glycoprotein in drug transport: A review. Drug Dev Res 2022;83(4):825-841 View Article PubMed/NCBI
  24. Mollazadeh S, Sahebkar A, Hadizadeh F, Behravan J, Arabzadeh S. Structural and functional aspects of P-glycoprotein and its inhibitors. Life Sci 2018;214:118-123 View Article PubMed/NCBI
  25. Marzolini C, Paus E, Buclin T, Kim RB. Polymorphisms in human MDR1 (P-glycoprotein): recent advances and clinical relevance. Clin Pharmacol Ther 2004;75(1):13-33 View Article PubMed/NCBI
  26. Ieiri I, Takane H, Hirota T, Otsubo K, Higuchi S. Genetic polymorphisms of drug transporters: pharmacokinetic and pharmacodynamic consequences in pharmacotherapy. Expert Opin Drug Metab Toxicol 2006;2(5):651-674 View Article PubMed/NCBI
  27. Yendapalli VSNR, Williams K, Oroszi T. Role of P-Gp in Treatment of Cancer. J Cancer Ther 2025;16(1):1-17 View Article
  28. Acuña-Guzman V, Montoya-Alfaro ME, Negrón-Ballarte LP, Solis-Calero C. A Machine Learning Approach for Predicting Caco-2 Cell Permeability in Natural Products from the Biodiversity in Peru. Pharmaceuticals (Basel) 2024;17(6):750 View Article PubMed/NCBI
  29. Kong X, Lin K, Wu G, Tao X, Zhai X, Lv L, et al. Machine Learning Techniques Applied to the Study of Drug Transporters. Molecules 2023;28(16):5936 View Article PubMed/NCBI
  30. Nies AT, Koepsell H, Damme K, Schwab M. Organic cation transporters (OCTs, MATEs), in vitro and in vivo evidence for the importance in drug therapy. In: Fromm M, Kim R (eds). Drug Transporters. Handbook of Experimental Pharmacology, vol 201. Berlin, Heidelberg: Springer Berlin Heidelberg; 2011:105-167 View Article PubMed/NCBI
  31. Chen Y, Li S, Brown C, Cheatham S, Castro RA, Leabman MK, et al. Effect of genetic variation in the organic cation transporter 2 on the renal elimination of metformin. Pharmacogenet Genomics 2009;19(7):497-504 View Article PubMed/NCBI
  32. Filipski KK, Loos WJ, Verweij J, Sparreboom A. Interaction of Cisplatin with the human organic cation transporter 2. Clin Cancer Res 2008;14(12):3875-3880 View Article PubMed/NCBI
  33. DeGorter MK, Xia CQ, Yang JJ, Kim RB. Drug transporters in drug efficacy and toxicity. Annu Rev Pharmacol Toxicol 2012;52:249-273 View Article PubMed/NCBI
  34. Caetano-Pinto P, Stahl SH. Renal Organic Anion Transporters 1 and 3 In Vitro: Gone but Not Forgotten. Int J Mol Sci 2023;24(20):15419 View Article PubMed/NCBI
  35. Prueksaritanont T, Chu X, Gibson C, Cui D, Yee KL, Ballard J, et al. Drug-drug interaction studies: regulatory guidance and an industry perspective. AAPS J 2013;15(3):629-645 View Article PubMed/NCBI
  36. Jain S, Ecker GF. In Silico Approaches to Predict Drug-Transporter Interaction Profiles: Data Mining, Model Generation, and Link to Cholestasis. Methods Mol Biol 2019;1981:383-396 View Article PubMed/NCBI
  37. Duan H, Lou C, Gu Y, Wang Y, Li W, Liu G, et al. In Silico prediction of inhibitors for multiple transporters via machine learning methods. Mol Inform 2024;43(3):e202300270 View Article PubMed/NCBI
  38. Schuetz JD, Swaan PW, Tweedie DJ. The role of transporters in toxicity and disease. Drug Metab Dispos 2014;42(4):541-545 View Article PubMed/NCBI
  39. Zhao M, Ma J, Li M, Zhang Y, Jiang B, Zhao X, et al. Cytochrome P450 Enzymes and Drug Metabolism in Humans. Int J Mol Sci 2021;22(23):12808 View Article PubMed/NCBI
  40. Srinivasan AV. Propranolol: A 50-Year Historical Perspective. Ann Indian Acad Neurol 2019;22(1):21-26 View Article PubMed/NCBI
  41. Gorski JC, Hall SD, Jones DR, VandenBranden M, Wrighton SA. Regioselective biotransformation of midazolam by members of the human cytochrome P450 3A (CYP3A) subfamily. Biochem Pharmacol 1994;47(9):1643-1653 View Article PubMed/NCBI
  42. Ingelman-Sundberg M. Pharmacogenetics of cytochrome P450 and its applications in drug therapy: the past, present and future. Trends Pharmacol Sci 2004;25(4):193-200 View Article PubMed/NCBI
  43. Dayyih WA, Al-Ani I, Hailat M, Alarman SM, Zakaraya Z, Assab MA, et al. Review of grapefruit juice-drugs interactions mediated by intestinal CYP3A4 inhibition. J Appl Pharm Sci 2024;14(5):59-68 View Article
  44. Tyzack JD, Kirchmair J. Computational methods and tools to predict cytochrome P450 metabolism for drug discovery. Chem Biol Drug Des 2019;93(4):377-386 View Article PubMed/NCBI
  45. Cruciani G, Carosati E, De Boeck B, Ethirajulu K, Mackie C, Howe T, et al. MetaSite: understanding metabolism in human cytochromes from the perspective of the chemist. J Med Chem 2005;48(22):6970-6979 View Article PubMed/NCBI
  46. Rydberg P, Gloriam DE, Zaretzki J, Breneman C, Olsen L. SMARTCyp: A 2D Method for Prediction of Cytochrome P450-Mediated Drug Metabolism. ACS Med Chem Lett 2010;1(3):96-100 View Article PubMed/NCBI
  47. Sonaji P, Subramanian L, Rajesh M. Artificial intelligence-driven drug interaction prediction. 2024;17(2):297-305 View Article
  48. Lai Y, Chu X, Di L, Gao W, Guo Y, Liu X, et al. Recent advances in the translation of drug metabolism and pharmacokinetics science for drug discovery and development. Acta Pharm Sin B 2022;12(6):2751-2777 View Article PubMed/NCBI
  49. Wu K, Kwon SH, Zhou X, Fuller C, Wang X, Vadgama J, et al. Overcoming Challenges in Small-Molecule Drug Bioavailability: A Review of Key Factors and Approaches. Int J Mol Sci 2024;25(23):13121 View Article PubMed/NCBI
  50. Polanski J. Receptor dependent multidimensional QSAR for modeling drug—receptor interactions. Curr Med Chem 2009;16(25):3243-3257 View Article PubMed/NCBI
  51. Komura H, Watanabe R, Mizuguchi K. The Trends and Future Prospective of In Silico Models from the Viewpoint of ADME Evaluation in Drug Discovery. Pharmaceutics 2023;15(11):2619 View Article PubMed/NCBI
  52. Lynch C, Sakamuru S, Ooka M, Huang R, Klumpp-Thomas C, Shinn P, et al. High-Throughput Screening to Advance In Vitro Toxicology: Accomplishments, Challenges, and Future Directions. Annu Rev Pharmacol Toxicol 2024;64:191-209 View Article PubMed/NCBI
  53. Dranchak PK, Oliphant E, Queme B, Lamy L, Wang Y, Huang R, et al. In vivo quantitative high-throughput screening for drug discovery and comparative toxicology. Dis Model Mech 2023;16(3):dmm049863 View Article PubMed/NCBI
  54. Dueñas ME, Peltier-Heap RE, Leveridge M, Annan RS, Büttner FH, Trost M. Advances in high-throughput mass spectrometry in drug discovery. EMBO Mol Med 2023;15(1):e14850 View Article PubMed/NCBI
  55. Chou WC, Lin Z. Machine learning and artificial intelligence in physiologically based pharmacokinetic modeling. Toxicol Sci 2023;191(1):1-14 View Article PubMed/NCBI
  56. Xiong G, Wu Z, Yi J, Fu L, Yang Z, Hsieh C, et al. ADMETlab 2.0: an integrated online platform for accurate and comprehensive predictions of ADMET properties. Nucleic Acids Res 2021;49(W1):W5-W14 View Article PubMed/NCBI
  57. Price E, Kalvass JC, DeGoey D, Hosmane B, Doktor S, Desino K. Global Analysis of Models for Predicting Human Absorption: QSAR, In Vitro, and Preclinical Models. J Med Chem 2021;64(13):9389-9403 View Article PubMed/NCBI
  58. Myung Y, de Sá AGC, Ascher DB. Deep-PK: deep learning for small molecule pharmacokinetic and toxicity prediction. Nucleic Acids Res 2024;52(W1):W469-W475 View Article PubMed/NCBI
  59. Cozac R, Hasic H, Choong JJ, Richard V, Beheshti L, Froehlich C, et al. kMoL: an open-source machine and federated learning library for drug discovery. J Cheminform 2025;17(1):22 View Article PubMed/NCBI
  60. Kojima R, Ishida S, Ohta M, Iwata H, Honma T, Okuno Y. kGCN: a graph-based deep learning framework for chemical structures. J Cheminform 2020;12(1):32 View Article PubMed/NCBI
  61. Li P, Hua L, Ma Z, Hu W, Liu Y, Zhu J. Conformalized Graph Learning for Molecular ADMET Property Prediction and Reliable Uncertainty Quantification. J Chem Inf Model 2024;64(23):8705-8717 View Article PubMed/NCBI
  62. Piazza I, Beaton N, Bruderer R, Knobloch T, Barbisan C, Chandat L, et al. A machine learning-based chemoproteomic approach to identify drug targets and binding sites in complex proteomes. Nat Commun 2020;11(1):4200 View Article PubMed/NCBI
  63. Heikamp K, Bajorath J. Support vector machines for drug discovery. Expert Opin Drug Discov 2014;9(1):93-104 View Article PubMed/NCBI
  64. Rodríguez-Pérez R, Bajorath J. Evolution of Support Vector Machine and Regression Modeling in Chemoinformatics and Drug Discovery. J Comput Aided Mol Des 2022;36(5):355-362 View Article PubMed/NCBI
  65. Sun R, Fei F, Wang M, Jiang J, Yang G, Yang N, et al. Integration of metabolomics and machine learning revealed tryptophan metabolites are sensitive biomarkers of pemetrexed efficacy in non-small cell lung cancer. Cancer Med 2023;12(18):19245-19259 View Article PubMed/NCBI
  66. Abramson J, Adler J, Dunger J, Evans R, Green T, Pritzel A, et al. Accurate structure prediction of biomolecular interactions with AlphaFold 3. Nature 2024;630(8016):493-500 View Article PubMed/NCBI
  67. Jumper J, Evans R, Pritzel A, Green T, Figurnov M, Ronneberger O, et al. Highly accurate protein structure prediction with AlphaFold. Nature 2021;596(7873):583-589 View Article PubMed/NCBI
  68. Tingle BI, Tang KG, Castanon M, Gutierrez JJ, Khurelbaatar M, Dandarchuluun C, et al. ZINC-22─A Free Multi-Billion-Scale Database of Tangible Compounds for Ligand Discovery. J Chem Inf Model 2023;63(4):1166-1176 View Article PubMed/NCBI
  69. Aria M, Cuccurullo C, Gnasso A. A comparison among interpretative proposals for Random Forests. Machine Learning with Applications 2021;6:100094 View Article
  70. Paul D, Sanap G, Shenoy S, Kalyane D, Kalia K, Tekade RK. Artificial intelligence in drug discovery and development. Drug Discov Today 2021;26(1):80-93 View Article PubMed/NCBI
  71. Zhou G, Rusnac DV, Park H, Canzani D, Nguyen HM, Stewart L, et al. An artificial intelligence accelerated virtual screening platform for drug discovery. Nat Commun 2024;15(1):7761 View Article
  72. Taherdoost H, Ghofrani A. AI’s role in revolutionizing personalized medicine by reshaping pharmacogenomics and drug therapy. Intelligent Pharmacy 2024;2(5):643-650 View Article
  73. Abdelhalim H, Berber A, Lodi M, Jain R, Nair A, Pappu A, et al. Artificial Intelligence, Healthcare, Clinical Genomics, and Pharmacogenomics Approaches in Precision Medicine. Front Genet 2022;13:929736 View Article PubMed/NCBI
  74. Alowais SA, Alghamdi SS, Alsuhebany N, Alqahtani T, Alshaya AI, Almohareb SN, et al. Revolutionizing healthcare: the role of artificial intelligence in clinical practice. BMC Med Educ 2023;23(1):689 View Article PubMed/NCBI
  75. Kim GB, Lee S, Kim H, Yang DH, Kim YH, Kyung YS, et al. Three-Dimensional Printing: Basic Principles and Applications in Medicine and Radiology. Korean J Radiol 2016;17(2):182-197 View Article PubMed/NCBI
  76. Peng H, Han B, Tong T, Jin X, Peng Y, Guo M, et al. 3D printing processes in precise drug delivery for personalized medicine. Biofabrication 2024;16(3):032001 View Article PubMed/NCBI
  77. Milliken RL, Quinten T, Andersen SK, Lamprou DA. Application of 3D printing in early phase development of pharmaceutical solid dosage forms. Int J Pharm 2024;653:123902 View Article PubMed/NCBI
  78. Öblom H, Cornett C, Bøtker J, Frokjaer S, Hansen H, Rades T, et al. Data-enriched edible pharmaceuticals (DEEP) of medical cannabis by inkjet printing. Int J Pharm 2020;589:119866 View Article PubMed/NCBI
  79. Vaz VM, Kumar L. 3D Printing as a Promising Tool in Personalized Medicine. AAPS PharmSciTech 2021;22(1):49 View Article PubMed/NCBI
  80. Goyanes A, Buanz AB, Basit AW, Gaisford S. Fused-filament 3D printing (3DP) for fabrication of tablets. Int J Pharm 2014;476(1-2):88-92 View Article PubMed/NCBI
  81. Katsiotis CS, Tikhomirov E, Leliopoulos C, Strømme M, Welch K. Development of a simple paste for 3D printing of drug formulations containing a mesoporous material loaded with a poorly water-soluble drug. Eur J Pharm Biopharm 2024;198:114270 View Article PubMed/NCBI
  82. Bernatoniene J, Stabrauskiene J, Kazlauskaite JA, Bernatonyte U, Kopustinskiene DM. The Future of Medicine: How 3D Printing Is Transforming Pharmaceuticals. Pharmaceutics 2025;17(3):390 View Article PubMed/NCBI
  83. Aina M, Baillon F, Sescousse R, Sanchez-Ballester NM, Begu S, Soulairol I, et al. From conception to consumption: Applications of semi-solid extrusion 3D printing in oral drug delivery. Int J Pharm 2025;674:125436 View Article PubMed/NCBI
  84. Iftekar SF, Aabid A, Amir A, Baig M. Advancements and Limitations in 3D Printing Materials and Technologies: A Critical Review. Polymers (Basel) 2023;15(11):2519 View Article PubMed/NCBI
  85. Elbadawi M, McCoubrey LE, Gavins FKH, Ong JJ, Goyanes A, Gaisford S, et al. Harnessing artificial intelligence for the next generation of 3D printed medicines. Adv Drug Deliv Rev 2021;175:113805 View Article PubMed/NCBI
  86. Patra JK, Das G, Fraceto LF, Campos EVR, Rodriguez-Torres MDP, Acosta-Torres LS, et al. Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnology 2018;16(1):71 View Article PubMed/NCBI
  87. Su S, Kang PM. Systemic Review of Biodegradable Nanomaterials in Nanomedicine. Nanomaterials (Basel) 2020;10(4):656 View Article PubMed/NCBI
  88. Das RP, Gandhi VV, Singh BG, Kunwar A. Passive and Active Drug Targeting: Role of Nanocarriers in Rational Design of Anticancer Formulations. Curr Pharm Des 2019;25(28):3034-3056 View Article PubMed/NCBI
  89. Man DK, Chow MY, Casettari L, Gonzalez-Juarrero M, Lam JK. Potential and development of inhaled RNAi therapeutics for the treatment of pulmonary tuberculosis. Adv Drug Deliv Rev 2016;102:21-32 View Article PubMed/NCBI
  90. Ojo AO, Morales JM, González-Molina M, Steffick DE, Luan FL, Merion RM, et al. Comparison of the long-term outcomes of kidney transplantation: USA versus Spain. Nephrol Dial Transplant 2013;28(1):213-220 View Article PubMed/NCBI
  91. Saneja A, Kumar R, Mintoo MJ, Dubey RD, Sangwan PL, Mondhe DM, et al. Gemcitabine and betulinic acid co-encapsulated PLGA-PEG polymer nanoparticles for improved efficacy of cancer chemotherapy. Mater Sci Eng C Mater Biol Appl 2019;98:764-771 View Article PubMed/NCBI
  92. Norori N, Hu Q, Aellen FM, Faraci FD, Tzovara A. Addressing bias in big data and AI for health care: A call for open science. Patterns (N Y) 2021;2(10):100347 View Article PubMed/NCBI
  93. Mirakhori F, Niazi SK. Harnessing the AI/ML in Drug and Biological Products Discovery and Development: The Regulatory Perspective. Pharmaceuticals (Basel) 2025;18(1):47 View Article PubMed/NCBI
  94. U.S. Food and Drug Administration. Emerging Technology Program (ETP). 2025. Available from: https://www.fda.gov/about-fda/center-drug-evaluation-and-research-cder/emerging-technology-program-etp. Accessed May 14, 2025
  95. Zagalo DM, Silva BMA, Silva C, Simões S, Sousa JJ. A quality by design (QbD) approach in pharmaceutical development of lipid-based nanosystems: A systematic review. J Drug Deliv Sci Technol 2022;70:103207 View Article
  96. International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use. Development and manufacture of drug substances (chemical entities and biotechnological/biological entities) Q11. 2012. Available from: https://database.ich.org/sites/default/files/Q11%20Guideline.pdf
  97. International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use. Pharmaceutical development Q8(R2). 2009. Available from: https://database.ich.org/sites/default/files/Q8_R2_Guideline.pdf
  98. Musuamba FT, Skottheim Rusten I, Lesage R, Russo G, Bursi R, Emili L, et al. Scientific and regulatory evaluation of mechanistic in silico drug and disease models in drug development: Building model credibility. CPT Pharmacometrics Syst Pharmacol 2021;10(8):804-825 View Article PubMed/NCBI
  99. Vamathevan J, Clark D, Czodrowski P, Dunham I, Ferran E, Lee G, et al. Applications of machine learning in drug discovery and development. Nat Rev Drug Discov 2019;18(6):463-477 View Article PubMed/NCBI

About this Article

Cite this article
Ali HB, Khan MB. Artificial Intelligence-driven Strategies and Modern Innovations to Surpass Biopharmaceutic Limitations in Traditional Drug Development. J Explor Res Pharmacol. 2025;10(3):e00025. doi: 10.14218/JERP.2025.00025.
Copy        Export to RIS        Export to EndNote
Article History
Received Revised Accepted Published
June 3, 2025 June 6, 2025 August 7, 2025 September 10, 2025
DOI http://dx.doi.org/10.14218/JERP.2025.00025
  • Journal of Exploratory Research in Pharmacology
  • pISSN 2993-5121
  • eISSN 2572-5505
Back to Top

Artificial Intelligence-driven Strategies and Modern Innovations to Surpass Biopharmaceutic Limitations in Traditional Drug Development

Haneen Badreldin Ali, Muhammad Burhan Khan
  • Reset Zoom
  • Download TIFF