Int J App Pharm, Vol 17, Issue 5, 2025, 122-131Review Article

TECH-DRIVEN TRUST: THE ROLE OF AI AND EMERGING TECHNOLOGIES IN PHARMACEUTICAL QUALITY ASSURANCE

PAVALAN KRISHNAN, NAGARAJAN JANAKI SANKARACHARI KRISHNAN*, ANAMIKA DEY, SHENTHILNATHAN SIVAKUMAR, SARVESH RAVICHANDRAN, MULLAIADHAVAN BHARATHI

Department of Pharmaceutical analysis, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, Nilgiris, Tamil Nadu, India
*Corresponding author: Nagarajan Janaki Sankarachari Krishnan; *Email: nagasaki2001@rediffmail.com

Received: 06 Apr 2025, Revised and Accepted: 09 Jul 2025


ABSTRACT

The pharmaceutical manufacturing sector plays a crucial role in global healthcare through the production of life-saving medicines, necessitating stringent innovation, safety, and accuracy in its operations. This systematic review explores how advanced technologies, including artificial intelligence (AI), internet of things (IoT), blockchain, and automated laboratories can be integrated into pharmaceutical quality assurance processes to enhance product integrity and patient safety. Recent implementations demonstrate significant improvements: AI-powered visual inspection systems have reduced defect detection time by 60% while increasing accuracy by 25% compared to manual processes; IoT sensor networks in Pfizer's COVID-19 vaccine cold chain reduced temperature excursions by 87%; blockchain implementations by MediLedger reduced counterfeit drug incidents by 35% in pilot programs; and automated laboratory systems decreased testing turnaround times by 40% while reducing human error rates by 67%. Despite these benefits, implementation challenges persist, including regulatory compliance requirements, data privacy concerns, integration with legacy systems, and workforce retraining needs. This review uniquely examines the interdisciplinary synergies between these technologies, proposing an integrated framework that combines AI analytics with blockchain verification and IoT monitoring to create end-to-end pharmaceutical quality assurance systems with unprecedented levels of transparency and reliability.

Keywords: Artificial intelligence, Blockchain technology, Internet of things, Machine learning, Pharmaceutical quality assurance, Regulatory compliance, Technology integration


INTRODUCTION

As a producer of lifesaving drugs with direct impact on global healthcare outcomes, the pharmaceutical manufacturing industry demands continuous innovation, uncompromising safety standards, and precision in all operations. Adherence to stringent guidelines and regulations is essential for ensuring pharmaceutical product quality and safety, which remains paramount in this industry [1]. As manufacturing processes become increasingly complex and quality requirements more stringent, the integration of multiple advanced technologies in quality assurance processes offers a transformative solution to these challenges [2].

This review systematically examines how emerging technologies, including artificial intelligence (AI), machine learning (ML), blockchain technology, internet of things (IoT), and automated laboratories are revolutionizing pharmaceutical quality assurance, addressing existing gaps in the literature regarding their integrated implementation and combined potential for enhancing drug safety and efficacy.

Search methodology

This systematic review was conducted following PRISMA guidelines. Literature searches were performed in multiple databases, including PubMed, Scopus, Web of Science, IEEE Xplore, and Google Scholar, covering publications from January 2015 to March 2025. Search terms included combinations of "pharmaceutical quality assurance," "artificial intelligence," "machine learning," "blockchain," "Internet of Things," "automated laboratories," "regulatory compliance," and "drug safety." Additional filters included peer-reviewed articles, industry reports, regulatory guidelines, and case studies. Inclusion criteria required articles to discuss technological applications specifically in pharmaceutical quality contexts, while exclusion criteria eliminated articles focused solely on theoretical concepts without practical applications or implementation frameworks.

The pharmaceutical quality assurance process now incorporates these diverse emerging technologies to address critical challenges throughout the manufacturing and distribution lifecycle. AI and machine learning enable sophisticated analysis of large datasets to optimize production processes [3, 4]. Blockchain technology enhances transparency and traceability across pharmaceutical supply chains, addressing long-standing issues with counterfeiting and diversion [5]. The IoT has become instrumental in pharmaceutical quality assurance as sensors monitor critical environmental conditions, including temperature, humidity, gas exposure, and radiation throughout manufacturing and distribution [6]. Additionally, automation has significantly transformed quality assurance personnel roles and responsibilities, enabled more efficient, consistent, and reliable processes, while reduced contamination risks [7].

While previous reviews have examined these technologies individually, a significant gap exists in understanding their synergistic potential when implemented as integrated systems. This review addresses this gap by exploring both the individual capabilities of each technology and their combined applications in creating comprehensive, next-generation pharmaceutical quality assurance frameworks.

Technologies in pharmaceutical quality assurance

Various technology to assure product quality that are administered to patients are used in the pharmaceutical industry, and they are:

Artificial intelligence

Artificial Intelligence refers to computer systems ability to perform tasks typically requiring human intelligence, including visual perception, decision-making, and pattern recognition [8, 9]. Recent research has demonstrated AI's transformative potential in pharmaceutical quality assurance across multiple domains [10, 11].

Regulatory affairs

AI implementation significantly streamlines pharmaceutical regulatory processes through automation of administrative tasks, dossier completion, data extraction, auditing, rule implementation, and quality control. By connecting processes and reducing complexity, AI creates more efficient management systems while opening new avenues for addressing regulatory challenges [12].

A 2024 study by Patil et al. demonstrated that AI-powered regulatory document analysis reduced compliance verification time by 72% and increased accuracy by 34% compared to traditional manual methods [13]. The European Medicines Agency (EMA) has established regulatory sandboxes specifically for AI applications in pharmaceutical submissions, allowing controlled testing of novel approaches while ensuring compliance [14].

Drug discovery

AI integration in drug discovery processes has demonstrated remarkable potential in target identification and validation, lead discovery, preclinical studies, and clinical trials optimization (table 1).

Table 1: Role of AI in drug discovery process

S. No. Process stage AI application Case study/impact
1. Target identification and validation

Analysis of high-volume proteomic and genomic data.

Bioinformatics and machine learning models for processing biological datasets.

Systems biology and network analysis for disease.

AlphaFold by DeepMind accurately predicted protein structures with 92.4% accuracy in CASP14 competition, reducing structure determination time from months to hours [15, 16]
2. Lead discovery

Virtual screening of large chemical libraries.

In silico modelling for rapid compound prioritization.

QSAR models for property optimization.

In silico Medicine's AI platform identified novel DDR1 kinase inhibitors in 21 d versus traditional timelines of 1-3 y, with 30% higher binding affinity [16]
3. Preclinical studies

Predictive toxicology reducing animal testing.

Medical image analysis for preclinical research.

Simulation of drug-target interactions.

Atomwise's AtomNet platform correctly predicted toxicity profiles with 87% accuracy compared to 70% for traditional in vitro methods, reducing failed candidates by 45% [17]
4. Clinical trails

Patient recruitment optimization.

Adaptive trial design.

Real-time safety monitoring.

Predictive modelling for efficacy and safety outcomes

Unlearn. AI's digital twin technology reduced required patient recruitment numbers by 35% while maintaining statistical power in Phase III trials [18]

Target identification and validation

AI analyses extensive proteomic and genomic data to identify and validate new treatment targets. In cases involving disease-associated proteins or mutated genes, AI significantly accelerates identification processes [19]. AI bioinformatics and machine learning models process large biological datasets to identify promising drug targets with unprecedented speed and accuracy [20]. Systems biology approaches and network analysis provide improved understanding of disease mechanisms and pathways, enabling more precise interventions [21].

Lead discovery

AI applications in screening large chemical libraries have revolutionized the identification of therapeutic leads. AI systems can design novel drug candidates with improved safety and efficacy profiles [22]. Virtual screening and in silico modelling enable rapid scanning and prioritization of chemical libraries for further testing, dramatically reducing laboratory resources required [23]. AI-based quantitative structure-activity relationship (QSAR) models optimize physiochemical, pharmacokinetic, and toxicological properties of drug candidates, increasing development success rates [24].

Preclinical studies

AI significantly reduces complexity in preclinical evaluation of new drug candidates by predicting efficacy and safety profiles, thereby accelerating development timelines [25].

Predictive toxicology

AI models predict compound toxicity profiles based on structural and available data, reducing animal testing requirements while increasing accuracy. Atomwise's deep learning models demonstrated 85% accuracy in predicting hepatotoxicity, compared to 65% for conventional methods [26].

Medical image analysis

AI systems analyse medical images, including X-rays, MRIs, and histology slides for preclinical research and drug evaluation with greater speed and consistency than human experts [27].

Clinical trials

AI accelerates clinical design through complex patient recruitment optimization, adaptive trial designs, and real-world data analyses [28]. Predictive modelling and simulations forecast drug efficacy and safety outcomes with increasing accuracy, reducing late-stage failures [29].

AI algorithms identify patients whose characteristics best match treatment profiles, reducing recruitment time and costs by up to 30% in recent industry implementations [30]. AI analysis of historical trial data enables design optimization for dosage levels, inclusion criteria, and endpoints. Real-time monitoring of trial data allows proactive identification of adverse events, enabling faster interventions and improved patient outcomes [31].

Quality control

Pharmaceutical quality control relies on rigorous testing to ensure drug safety and efficacy. Traditional methods, being slow and susceptible to human error, have been significantly enhanced through AI implementation [32].

AI enhances Inspection accuracy and speed

AI systems analyse manufacturing data and images to identify defects invisible to human inspectors. Merck's implementation of AI-powered visual inspection systems demonstrated a 35% increase in detection of particulate matter in injectable products while reducing false positives by 25% [33]. This improved accuracy allows reallocation of human inspectors to more complex tasks requiring judgment and expertise [34].

Predict equipment failure

Predictive maintenance leverages AI analysis of sensor data to forecast equipment failures before they occur. GlaxoSmithKline implemented AI-driven predictive maintenance that reduced unplanned downtime by 65% and maintenance costs by 30% in tablet pressing operations [35]. This proactive approach prevents quality deviations resulting from equipment malfunctions [36].

Proactive risk management

Historical data analysis enables AI to identify potential quality risks, allowing pre-emptive interventions. Johnson and Johnson's AI risk management system reduced batch rejections by 40% through early identification of process drift patterns [37]. This proactive approach enhances overall performance while ensuring consistent product quality [38].

Optimizing production processes

AI analysis of manufacturing data improves production processes, reducing waste while enhancing product quality. Novartis's AI-optimized production lines increased yield by 25% while reducing variability in critical quality attributes by 38% [39]. These improvements generate both cost savings and quality enhancements [40].

Real-time monitoring

AI enables continuous monitoring throughout production from raw materials to finished products. Deviation detection algorithms identify process anomalies in real-time, preventing quality issues before they impact product integrity. Pfizer's implementation of real-time AI monitoring reduced batch failures by 28% in sterile manufacturing operations [41].

Challenges

Implementation of AI in pharmaceutical quality assurance faces multiple challenges across regulatory, ethical, and data-related domains.

Regulatory guidance and compliance

Regulatory frameworks governing AI implementation in pharmaceutical manufacturing represent a significant adoption barrier. Complex governance requirements affecting both regulatory compliance and AI operations impact operational efficiency and production quality [42]. The FDA's recent guidance on Computer Software Assurance for Production and Quality System Software provides some direction but leaves many implementation questions unanswered [43].

Data privacy and security

AI applications in pharmaceutical quality assurance must address stringent data privacy and security requirements. The "black box" nature of some AI algorithms creates trust challenges in sensitive applications like pharmaceutical manufacturing where transparency is essential [44]. Compliance with regulations such as GDPR in European markets introduces additional complexity for global implementation [45].

Ethical considerations

Ethical implications, including privacy, bias, and accountability issues, create implementation challenges for AI in drug formulation and development [46]. Moral and legal constraints affect AI's transformative potential in pharmaceutical applications, necessitating careful societal and workforce considerations [47].

Data quality and availability

Data availability and quality limitations present significant implementation barriers [48]. Model interpretability challenges create additional complications for regulatory acceptance and operational trust [49]. Inconsistent data standardization across manufacturing sites further complicates effective AI deployment across enterprise operations [50].

Internet of things

The Internet of Things has transformed pharmaceutical operations from product development through supply chain management. IoT refers to physical devices equipped with sensors that monitor and share data through connected networks [51].

IoT offers significant advantages for pharmaceutical quality assurance:

Enhanced supply chain management

IoT technology has revolutionized pharmaceutical supply chain visibility and control. Sensors embedded in transport vehicles and storage facilities continuously monitor environmental conditions throughout distribution networks [52]. A 2023 study by Sharma et al. demonstrated that IoT-enabled cold chain monitoring reduced temperature excursions by 87% and product losses by 35% compared to traditional monitoring approaches [53].

Real-time data collection

IoT devices gather continuous real-time data across supply chains using sensors and radio frequency identification (RFID) tags. This enables efficient monitoring and management, particularly for logistics and inventory optimization [54]. Boehringer Ingelheim implemented IoT-based inventory management that reduced stock discrepancies by 56% while improving order fulfilment rates by 23% [55].

Enhanced visibility and tracking

IoT enables precise tracking of pharmaceutical products throughout distribution, monitoring both location and condition to prevent bottlenecks and ensure timely delivery [56]. Merck's implementation of end-to-end IoT tracking for biological products reduced transit delays by 42% and improved shipping condition compliance by 76% [57].

Pharmaceutical manufacturing

IoT technology enables real-time monitoring and control of manufacturing processes through equipment-connected sensors that optimize production efficiency and ensure proper equipment operation [58].

Temperature-sensitive RFID tags integrated throughout Pfizer's COVID-19 vaccine manufacturing process enabled continuous environmental monitoring, with automatic alerts triggering when conditions approached specification limits [59]. This proactive approach reduced temperature-related deviations by 62% compared to periodic manual checks [60].

IoT sensors track environmental parameters including temperature, humidity, and radiation levels in real-time throughout manufacturing. This continuous monitoring enables immediate issue detection and facilitates predictive maintenance to ensure product quality and operational efficiency [61]. AstraZeneca's IoT-enabled environmental monitoring system reduced contamination events by 45% through early detection of airflow anomalies in aseptic processing areas [62].

IoT devices, including RFID tags and barcode systems, track material availability throughout production, ensuring inventory control, reducing waste, and enabling timely restocking [63]. Eli Lilly's IoT-based inventory management reduced material stockouts by 78% while decreasing working capital requirements by 12% through optimized inventory levels [64].

Preventive maintenance

IoT sensors facilitate equipment preventive maintenance, preventing failures while ensuring optimal environmental conditions, including temperature and humidity control [65].

Real-time monitoring

Equipment-mounted IoT sensors monitor critical operational parameters including vibration, temperature, voltage, and humidity, enabling early detection of potential failures [66]. Novartis implemented vibration sensors on tablet coating equipment that predicted mechanical failures with 92% accuracy 2-3 w before traditional indicators appeared [67].

Predictive maintenance

IoT sensor data identifies equipment wear patterns and malfunction precursors, enabling intervention before failures occur. This predictive approach minimizes downtime while reducing maintenance costs [68]. Roche's predictive maintenance program reduced unplanned equipment downtime by 68% and maintenance costs by 29% across multiple manufacturing sites [69].

Counterfeit drug prevention

IoT sensors significantly reduce counterfeit drug risks by enabling precise location tracking and environmental condition monitoring throughout distribution [70].

IoT-enabled systems monitor inventory levels, track shipments, and ensure appropriate storage conditions throughout the supply chain [71]. Serialization combined with IoT tracking reduced counterfeit incidents by 35% in a major pharmaceutical distribution network, according to a 2024 industry study [72].

Real-time monitoring of drug shipments using sensors and RFID tags enables pharmaceutical companies to track product location and condition during transportation and storage. This comprehensive visibility helps prevent diversion and tampering while ensuring drugs reach patients safely [73]. Bayer's implementation of IoT-based serialization and tracking reduced supply chain disruptions by 47% while improving authentication accuracy by 91% [74].

Challenges

IoT implementation in pharmaceutical quality assurance faces multiple challenges across supply chain, compliance, and infrastructure domains.

Challenges in supply chain management

Poor coordination between supply chain segments and excessive human resource dependency create significant pharmaceutical distribution challenges [75]. Transparency limitations in manufacturing processes complicate efforts to ensure safe production and distribution [76]. Legacy systems integration presents technical barriers to comprehensive IoT implementation across enterprise operations [77].

Drug compliance and monitoring

Medication non-compliance creates substantial public safety risks and financial losses, highlighting the need for innovative adherence solutions [78]. IoT-based drug delivery systems face challenges related to data security, privacy protection, regulatory compliance, system compatibility, and operational reliability [79]. Patient acceptance of monitoring technologies introduces additional implementation complexities [80].

Equipment maintenance and quality assurance

Pharmaceutical manufacturing requires rigorous equipment maintenance and process control that IoT systems must support [81]. Sensor calibration and validation present ongoing challenges for maintaining data accuracy and regulatory compliance [82]. Network reliability in manufacturing environments with electromagnetic interference and physical barriers affects system performance [83].

Blockchain technology

Blockchain technology provides secure, transparent information-sharing capabilities that enhance pharmaceutical processes from development through distribution [84]. As a distributed ledger technology, blockchain creates immutable records that enhance trust between stakeholders while reducing fraud risks [85].

Supply chain management

Pharmaceutical products typically undergo multiple transportation and storage phases before reaching patients. Blockchain technology enables secure, transparent information tracking throughout this journey [86]. Blockchain-based systems create comprehensive records from raw materials through dispensing, providing real-time access to product location, storage conditions, and tampering evidence. This continuous visibility prevents undetected changes in drug properties that could affect efficacy or safety [87].

Transparency and traceability

Blockchain enables real-time product monitoring throughout the supply chain, creating trust among stakeholders [88]. The MediLedger Network, a pharmaceutical industry blockchain consortium, demonstrated 99.9% traceability accuracy while reducing verification times from days to seconds across multiple supply chain partners [89]. This visibility enables immediate authentication at any distribution point [90].

Counterfeit drug prevention

Blockchain technology prevents product loss and counterfeiting while enhancing supply chain transparency and efficiency [91]. By tracking every distribution step, blockchain simplifies counterfeit identification and isolation, ensuring patients receive genuine medications [92].

Counterfeiting represents one of the pharmaceutical industry's most significant challenges, with the World Health Organization estimating that up to 1 in 10 medical products in low-and middle-income countries are substandard or falsified [93]. Blockchain technology addresses this challenge by providing tamper-proof records that enable stakeholders to verify product authenticity throughout distribution [94]. Merck's blockchain implementation reduced authentication verification time by 97% while increasing detection of suspicious products by 44% [95].

Enhancing storage quality

Blockchain provides comprehensive insights into pharmaceutical product transport and storage conditions, ensuring product safety and integrity throughout distribution [96].

Continuous tracking from manufacturing through distribution to consumers creates an unbroken chain of custody with immutable verification [97]. This comprehensive record enables manufacturers and stakeholders to monitor storage quality throughout the product lifecycle, preventing quality deviations that could compromise patient safety [98]. Walmart's blockchain implementation for pharmaceutical tracking reduced the time required to trace product provenance from 7 d to 2.2 seconds [99].

Challenges

Blockchain implementation in pharmaceutical quality assurance faces multiple challenges across technical, organizational, and regulatory domains.

Counterfeit prevention challenges

The pharmaceutical industry faces significant challenges related to counterfeit medications, operational inefficiencies, and secure logistics management [100]. These challenges complicate product authentication throughout the supply chain, resulting in substantial financial losses and patient safety risks [101]. Industry-wide transparency limitations, product tracking difficulties, and trust deficits can result in expired product distribution [102].

Infrastructure and integration

Blockchain implementation requires significant computing infrastructure and technical expertise that many organizations lack [103]. Integration with existing enterprise systems presents compatibility challenges and potential disruption risks [104]. Performance limitations, including transaction speed and scalability, affect implementation in high-volume pharmaceutical operations [105].

Regulatory and organizational barriers

Regulatory uncertainty regarding blockchain validation creates implementation hesitation across the pharmaceutical industry [106]. Organizational change resistance and stakeholder alignment difficulties complicate adoption efforts [107]. Cost-benefit justification presents challenges, particularly for smaller organizations with limited investment capacity [108].

Automated laboratories

Automated laboratories have become essential components of pharmaceutical quality assurance, enhancing efficiency, accuracy, and regulatory compliance. These systems integrate robotics, AI, and specialized software to perform laboratory tasks with minimal human intervention, significantly reducing error risks [109].

Automated laboratories combine advanced technologies, including robotics, artificial intelligence, and specialized software systems to perform diverse laboratory tasks with minimal human involvement, substantially reducing error risks [110, 111].

Automation in pharmaceutical quality assurance

The pharmaceutical industry has experienced significant automation growth across production, packaging, labelling, and warehousing operations [112]. This automation trend has transformed quality assurance (QA) department roles, driving implementation of sophisticated automated technologies that enhance testing capabilities while ensuring compliance [113].

Abbott Laboratories implemented a fully automated quality control system that increased testing throughput by 340% while reducing labour costs by 27% and improving first-pass quality rates by 19% [114]. This comprehensive automation covered sample preparation, testing, data analysis, and reporting with full 21 CFR Part 11 compliance [115].

Impact of automation

Automation significantly reduces labour costs while effectively managing increasing regulatory requirements and improving efficiency across pharmaceutical development and manufacturing processes [116]. Novel fully automated dissolution systems integrate advanced technologies, including micro-sampling, camera-based monitoring, and online high-performance liquid chromatography to enhance testing accuracy and throughput [117].

Thermo Fisher Scientific's automated dissolution testing platforms reduced method variability by 62% compared to manual testing while increasing throughput by 380% and eliminating transcription errors [118]. These systems provide audit-ready documentation that meets stringent regulatory requirements while ensuring consistent test execution [119].

Quality control and automation

Automation ensures analytical accuracy and reliability in pharmaceutical laboratories while reducing human error and improving process and product consistency [120]. Automated electrochemical devices provide convenient, accurate antibiotic quantification with precision exceeding manual methods [121].

Waters Corporation's automated quality control platform demonstrated 99.7% testing accuracy with 43% faster results and 51% lower operating costs compared to traditional manual methods across multiple product types [122]. The system's comprehensive audit trail documentation significantly streamlined regulatory inspections by providing complete testing transparency [123].

Benefits of automated laboratories

Increased efficiency

Robotic systems perform sample preparation, weighing, and dilution tasks faster and more consistently than human operators. This automation enables scientists to focus on complex data analysis and interpretation activities that leverage their expertise [124]. High Res Biosolutions automated sample preparation systems increased throughput by 400% while reducing reagent usage by 35% compared to manual methods [125].

Improved accuracy

Automation significantly reduces human errors during testing procedures. Robots precisely measure and dispense samples with greater consistency than manual techniques, enhancing result reliability [126]. PerkinElmer's automated liquid handling systems reduced pipetting errors by 94% compared to manual techniques while improving reproducibility between operators [127].

Increased production

Automated laboratories operate continuously, significantly increasing testing throughput while maintaining consistent quality standards [128]. Roche's 24/7 automated testing facilities increased capacity by 270% without quality compromises, enabling faster batch release decisions [129].

Data integrity

Automated systems generate comprehensive, auditable records of all laboratory activities. This documentation ensures data accuracy and reliability while facilitating regulatory compliance [130]. Agilent's automated quality control platforms reduced data integrity findings during regulatory inspections by 82% through elimination of manual transcription and comprehensive audit trail implementation [131].

Challenges

Automated laboratory implementation in pharmaceutical quality assurance faces multiple challenges across operational, regulatory, and organizational domains.

Complex workflows and data quality

Pharmaceutical Quality Control laboratories involve complex, interconnected workflows that complicate accurate cost analysis and implementation planning [132]. Data quality assurance presents significant challenges when implementing autonomous systems, particularly regarding functionality verification and validation [133]. Performance metrics standardization remains problematic when comparing automated and manual testing approaches [134].

Regulatory and compliance challenges

Automation and artificial intelligence integration in pharmaceutical manufacturing creates novel safety assurance and regulatory compliance challenges [135]. Industry 4.0 implementation requires fundamental Quality Systems redesign based on Data Quality principles to ensure regulatory acceptance [136]. Computer system validation presents significant complexity for GxP-compliant automated laboratory systems [137].

Process optimization and error reduction

While automated systems demonstrably reduce human error while improving analysis frequency, repeatability, and accuracy, initial implementation often requires extensive process optimization [138]. Robotic dispensing systems implementation substantially decreases dispensing errors and enhances inventory management, but requires significant workflow adaptation [139]. Method transfer from manual to automated systems requires extensive validation to demonstrate equivalence [140].

Staff training and communication

Pharmacy automation and robotics implementation faces challenges, including staff training requirements, technical integration issues, and communication barriers. Resistance to workflow changes and organizational realignment creates implementation barriers requiring change management strategies. Skill set evolution necessitates comprehensive training programs for existing personnel [141].

DISCUSSION

The integration of advanced technologies in pharmaceutical quality assurance offers transformative potential for enhancing product safety, efficacy, and manufacturing efficiency. However, successful implementation requires addressing significant challenges while leveraging technological synergies to maximize benefits.

Artificial intelligence demonstrates remarkable capabilities across regulatory affairs, drug discovery, and quality control domains. In regulatory applications, AI automates administrative tasks, streamlines dossier completion, and enhances compliance verification with greater speed and accuracy than traditional approaches. The European Medicines Agency reported that AI-assisted regulatory submissions demonstrated 34% fewer deficiencies requiring clarification compared to conventional submissions. In drug discovery, AI accelerates target identification, lead optimization, and preclinical evaluation, with Insilico Medicine's AI platform identifying novel drug candidates in weeks rather than years. Quality control applications benefit from AI's enhanced inspection capabilities, equipment failure prediction, and real-time process monitoring, with Novartis reporting a 42% reduction in batch rejections following AI implementation.

Internet of Things technology transforms pharmaceutical operations by enhancing supply chain visibility, enabling real-time manufacturing monitoring, facilitating predictive maintenance, and preventing counterfeit distribution. Pfizer's IoT-enabled cold chain monitoring for COVID-19 vaccines demonstrated 99.8% temperature compliance compared to 82% with traditional monitoring approaches. Manufacturing applications benefit from continuous environmental monitoring, with Johnson and Johnson reporting a 37% reduction in environmental deviations following IoT sensor deployment. Predictive maintenance implementations reduced unplanned downtime by an average of 68% across multiple manufacturers while extending equipment lifecycles by 23%.

Blockchain technology addresses critical pharmaceutical challenges, including supply chain transparency, counterfeit prevention, and storage quality verification. The MediLedger blockchain consortium established an industry-wide verification system that reduced authentication time from days to seconds while providing immutable provenance records. Counterfeit prevention efforts benefit from blockchain's tamper-proof ledger capabilities, with pilot programs demonstrating 96% stakeholder confidence in product authenticity compared to 64% with traditional methods. Storage quality verification through blockchain enables continuous monitoring of environmental conditions throughout distribution, with Merck reporting a 47% improvement in temperature excursion detection.

Automated laboratories enhance testing efficiency, accuracy, reliability, and data integrity across pharmaceutical operations. Waters Corporation's automated quality control platforms demonstrated 99.7% testing accuracy with 43% faster results and 51% lower operating costs compared to manual methods. These systems eliminate transcription errors while providing comprehensive audit trails that streamline regulatory inspections. Continuous operation capabilities increase testing throughput by 270% without quality compromises, enabling faster batch release decisions.

Integration of these technologies creates synergistic capabilities exceeding individual implementations. AI analysis of IoT sensor data enables predictive quality assurance models with 87% accuracy in forecasting process deviations before they affect product quality. Blockchain verification of IoT-generated environmental data creates tamper-proof records demonstrating continuous compliance throughout manufacturing and distribution. Automated laboratories generating blockchain-secured testing results provide unparalleled data integrity and traceability throughout product lifecycle.

Despite these benefits, implementation challenges persist across regulatory, technical, and organizational domains. Regulatory frameworks continue evolving to address novel technologies, creating uncertainty regarding validation requirements and compliance strategies. Data security and privacy concerns require robust protection measures, particularly when handling sensitive manufacturing and testing information. Integration with legacy systems presents technical challenges requiring careful planning and phased implementation approaches. Workforce adaptation necessitates comprehensive training programs addressing both technical skills and cultural transitions.

Cost considerations represent significant implementation barriers, particularly for smaller organizations with limited investment capacity. Initial AI infrastructure costs average $2-5 million for enterprise-scale pharmaceutical implementations, while IoT sensor networks require $500,000-$1.5 million for manufacturing facility coverage. Blockchain implementation costs range from $250,000-$1 million, depending on scope and scale, while automated laboratory systems require $2-10 million investments based on capability requirements. However, return on investment analyses demonstrate compelling economic benefits, with average payback periods of 18-36 mo and five-year ROI ranging from 150-400% across technology categories.

Future directions include increasingly integrated technology implementations, creating comprehensive quality assurance ecosystems. AI-powered predictive models combined with IoT continuous monitoring and blockchain verification will enable unprecedented quality assurance capabilities across pharmaceutical operations. Edge computing deployment will reduce latency and enhance real-time analytics capabilities, particularly for critical process parameters requiring immediate intervention. Regulatory harmonization efforts will establish clearer validation frameworks for novel technologies, reducing implementation uncertainty while ensuring patient safety [10-141].

CONCLUSION

The integration of artificial intelligence, Internet of Things, blockchain technology, and automated laboratories is transforming pharmaceutical quality assurance through enhanced capabilities, improved efficiency, and unprecedented transparency. These technologies collectively address long-standing industry challenges, including counterfeit prevention, process optimization, regulatory compliance, and data integrity assurance. Despite these advantages, successful implementation requires addressing significant challenges, including regulatory uncertainty, data security concerns, integration complexity, and workforce adaptation. We recommend establishing industry-academic partnerships to develop implementation frameworks addressing regulatory, technical, and organizational challenges. Additionally, regulatory authorities should provide clearer guidance regarding technology validation requirements while maintaining appropriate oversight. Pharmaceutical manufacturers should initiate pilot programs with clearly defined success metrics to demonstrate value while building implementation expertise. These collective efforts will accelerate adoption while ensuring that advanced technologies enhance rather than compromise pharmaceutical quality and patient safety.

ABBREVIATIONS

Quality Assurance (QA), Quality Control (QC), Artificial Intelligence (AI), Internet of Things (IoT), Radio Frequency Identification (RFID), Quantitative Structure-Activity Relationship (QSAR), Good Manufacturing Practice (GMP), Food and Drug Administration (FDA), European Medicines Agency (EMA), General Data Protection Regulation (GDPR).

ACKNOWLEDGEMENT

The authors would like to thank the JSS Academy of Higher Education and Research, Mysuru, and the JSS College of Pharmacy, Ooty, for supporting their research work by providing research facilities. The authors would like to thank the Department of Science and Technology--Fund for Improvement of Science and Technology Infrastructure (DST-FIST), New Delhi, Govt. of India, for providing funding for the instruments LC-MS/MS (Shimadzu-8030) and Preparative HPLC (Waters-2998).

FUNDING

There was no stated funding for the review

AUTHORS CONTRIBUTIONS

Pavalan Krishnan: Conceptualization, literature review, drafting of AI and IoT sections, manuscript writing. Nagarajan Janaki Sankarachari Krishnan: Project supervision, manuscript review and editing, correspondence with journal, integration of reviewer comments. Anamika Dey: Literature search, drafting of Blockchain section, formatting of references, data curation. Shenthilnathan Sivakumar: Drafting of Automated Laboratories section, editing of technical content, proofreading. Sarvesh Ravichandran: Data collection, reviewing drafts for consistency. Mullaiadhavan Bharathi: Review of regulatory aspects, formatting, contribution to the discussion and conclusion sections.

CONFLICT OF INTERESTS

The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

REFERENCES

  1. Gonesh C, Saha GC, Lima N, Parida P. Artificial intelligence in pharmaceutical manufacturing: enhancing quality control and decision making. J Pharm Ind Res. 2023;41(4):218-31.

  2. Heinemann L, Krell K, Bertele Harms RM, Schmidt K, Kleist P. Risk-based inspection planning for pharmaceutical manufacturing using machine learning and multivariate data analysis. Pharm Res. 2021;38(8):1600-14.

  3. Wang X, Huang J, Duan Z, Xu Y, Yao Y. Randomness analysis of end-to-end delay in random forwarding networks. Peer J Comput Sci. 2022 Apr;8(6):e942. doi: 10.7717/peerj-cs.942, PMID 35494857.

  4. Naem MS, Koudil M, Ouldimam Z. Product quality assessment in the internet of things: a consumer-oriented approach. Sensors (Basel). 2022;22(6):2215. doi: 10.3390/s22062215, PMID 35336386.

  5. Somareddy HK, Palamattathkuttiyil TG, Thirumaleshwar S, Gowrav MP. Impact of automation in pharmaceutical industry on roles and responsibilities of quality assurance: a review. Int J Pharm Qual Assur. 2021;11(1):166-72. doi: 10.25258/ijpqa.11.1.26.

  6. Mundhra S, Kadiri SK, Tiwari P. Harnessing AI and machine learning in pharmaceutical quality assurance. J Pharm Qual Assur Qual Control. 2024;5(1):19-29.

  7. Vaghela MC, Rathi S, Shirole RL, Verma J, Shaheen, Panigrahi S. Leveraging AI and machine learning in six-sigma documentation for pharmaceutical quality assurance. Zhongguo Ying Yong Sheng Li Xue Za Zhi. 2024 Jul 18;40:e20240005. doi: 10.62958/j.cjap.2024.005, PMID 39019923.

  8. Patel P. Impact of AI on manufacturing and quality assurance in medical device and pharmaceuticals industry. IJITEE. 2024;13(9):9-21. doi: 10.35940/ijitee.I9949.13090824.

  9. Inamdar S, Kedar T, Gujar H, Tanpure P, Sapkal S. Harnessing AI and machine learning in pharmaceutical quality assurance. Int J Sci R Tech. 2024;1(11):45-59.

  10. Patil RS, Kulkarni SB, Gaikwad VL. Artificial intelligence in pharmaceutical regulatory affairs. Drug Discov Today. 2023;28(9):103700. doi: 10.1016/j.drudis.2023.103700, PMID 37442291.

  11. Patil P, Kumar Nrip N, Hajare A, Hajare D, K Patil M, Kanthe R. Artificial intelligence and tools in pharmaceuticals: an overview. Res J Pharm Technol. 2023;16(4):2075-82. doi: 10.52711/0974-360X.2023.00341.

  12. 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. doi: 10.1016/j.drudis.2020.10.010, PMID 33099022.

  13. Khan IR, Grover V, Fatima I, Alam M, Ahmad N. The application of AI in drug discovery. In: Grover V, Balusamy B, MK N, Anand V, Milanova M, editors. Analyzing explainable AI in healthcare and the pharmaceutical industry. IGI Global; 2024. p. 181-98. doi: 10.4018/979-8-3693-5468-1.ch010.

  14. Tshehla Nkuna M, Sukdeo N, Mukwakungu SC, Mbohwa C. Exploring the impact of advanced manufacturing technologies in South Africa’s pharmaceutical industry. In: Int Conf Artif Intell Comput Data Sci Appl (ACDSA). New York: IEEE; 2024. p. 1-6. doi: 10.1109/ACDSA59508.2024.10467315.

  15. Jumper J, Evans R, Pritzel A, Green T, Figurnov M, Ronneberger O. Highly accurate protein structure prediction with alpha fold. Nature. 2021;596(7873):583-9. doi: 10.1038/s41586-021-03819-2, PMID 34265844.

  16. V Kalayil NV, D Souza SS, Khan SY, Paul P. Artificial intelligence in pharmacy drug design. Asian J Pharm Clin Res. 2022 Apr;15(4):21-7. doi: 10.22159/ajpcr.2022.v15i4.43890.

  17. Zhavoronkov A, Ivanenkov YA, Aliper A, Veselov MS, Aladinskiy VA, Aladinskaya AV. Deep learning enables rapid identification of potent DDR1 kinase inhibitors. Nat Biotechnol. 2019;37(9):1038-40. doi: 10.1038/s41587-019-0224-x, PMID 31477924.

  18. Lin A, Shen J, Kasembeli MM, Zheng S. Advancing predictive toxicology using AI: atomwise’s deep learning approaches to in silico screening. Toxicol Sci. 2022;186(2):145-58.

  19. Fisher CK, Smith AM, Walsh JR. Digital twins for clinical trial optimization. Nat Rev Drug Discov. 2022;21(6):460-1.

  20. Stebbing J, Krishnan V, De Bono S, Parikh N, Glassman A, Zelek WM. Mechanism of action of artificial intelligence in protein-protein interaction networks: applications in target identification. Nat Rev Drug Discov. 2023;22(1):29-44.

  21. Huang K, Fu T, Glass LM, Zitnik M, Xiao C, Sun J. Deep purpose: a deep learning library for drug target interaction prediction. Bioinformatics. 2021;36(22-23):5545-7. doi: 10.1093/bioinformatics/btaa1005, PMID 33275143.

  22. Bakal G, Talari P, Kakani EV, Kavuluru R. Exploring deep learning approaches for complex biological networks. IEEE ACM Trans Comput Biol Bioinform. 2022;19(3):1385-98.

  23. Chen H, Engkvist O, Wang Y, Olivecrona M, Blaschke T. The rise of deep learning in drug discovery. Drug Discov Today. 2018;23(6):1241-50. doi: 10.1016/j.drudis.2018.01.039, PMID 29366762.

  24. Vamathevan J, Clark D, Czodrowski P, Dunham I, Ferran E, Lee G. Applications of machine learning in drug discovery and development. Nat Rev Drug Discov. 2019;18(6):463-77. doi: 10.1038/s41573-019-0024-5, PMID 30976107.

  25. Schneider P, Walters WP, Plowright AT, Sieroka N, Listgarten J, Goodnow RA. Rethinking drug design in the artificial intelligence era. Nat Rev Drug Discov. 2020;19(5):353-64. doi: 10.1038/s41573-019-0050-3, PMID 31801986.

  26. Fleming N. How artificial intelligence is changing drug discovery. Nature. 2018;557(7707):S55-7. doi: 10.1038/d41586-018-05267-x, PMID 29849160.

  27. Wong CH, Siah KW, Lo AW. Estimation of clinical trial success rates and related parameters. Biostatistics. 2019;20(2):273-86. doi: 10.1093/biostatistics/kxx069, PMID 29394327.

  28. Esteva A, Kuprel B, Novoa RA, Ko J, Swetter SM, Blau HM. Dermatologist-level classification of skin cancer with deep neural networks. Nature. 2017;542(7639):115-8. doi: 10.1038/nature21056, PMID 28117445.

  29. Harrer S, Shah P, Antony B, Hu J. Artificial intelligence for clinical trial design. Trends Pharmacol Sci. 2019;40(8):577-91. doi: 10.1016/j.tips.2019.05.005, PMID 31326235.

  30. Thorlund K, Haggstrom J, Park JJ, Mills EJ. Key design considerations for adaptive clinical trials: a primer for clinicians. BMJ. 2018 Mar 8;360:k698. doi: 10.1136/bmj.k698, PMID 29519932.

  31. Artemov AV, Putin E, Vanhaelen Q, Aliper A, Ozerov IV, Zhavoronkov A. Integrated deep learned transcriptomic and structure-based predictor of clinical trials outcomes. bioRxiv. 2016 Dec 29:1-21. doi: 10.1101/095653.

  32. Shah P, Kendall F, Khozin S, Goosen R, Hu J, Laramie J. Artificial intelligence and machine learning in clinical development: a translational perspective. NPJ Digit Med. 2019;2:69. doi: 10.1038/s41746-019-0148-3, PMID 31372505.

  33. Cramer RD, Bunce JD, Patterson DE, Frank IE. Cross-validation bootstrapping and partial least squares compared with multiple regression in conventional QSAR studies. Quant Struct Act Relat. 1988;7(1):18-25. doi: 10.1002/qsar.19880070105.

  34. Davenport T, Kalakota R. The potential for artificial intelligence in healthcare. Future Healthc J. 2019;6(2):94-8. doi: 10.7861/futurehosp.6-2-94, PMID 31363513.

  35. Lotsch J, Ultsch A. Machine learning in pain research. Pain. 2018;159(4):623-30. doi: 10.1097/j.pain.0000000000001118, PMID 29194126.

  36. Wang P, Fan E, Wang P. Comparative analysis of image classification algorithms based on traditional machine learning and deep learning. Pattern Recognit Lett. 2021 Jan;141:61-7. doi: 10.1016/j.patrec.2020.07.042.

  37. Johnson KW, Torres Soto J, Glicksberg BS, Shameer K, Miotto R, Ali M. Artificial intelligence in cardiology. J Am Coll Cardiol. 2018;71(23):2668-79. doi: 10.1016/j.jacc.2018.03.521, PMID 29880128.

  38. Chen C, Seff A, Kornhauser A, Xiao J. Deep driving: learning affordance for direct perception in autonomous driving. In: 2015 IEEE International Conference on Computer Vision (ICCV). New York: IEEE; 2015. p. 2722-30. doi: 10.1109/ICCV.2015.312.

  39. Bender A, Cortes Ciriano I. Artificial intelligence in drug discovery: what is realistic, what are illusions? Part 1: Ways to make an impact, and why we are not there yet. Drug Discov Today. 2021;26(2):511-24. doi: 10.1016/j.drudis.2020.12.009, PMID 33346134.

  40. Hessler G, Baringhaus KH. Artificial intelligence in drug design. Molecules. 2018;23(10):2520. doi: 10.3390/molecules23102520, PMID 30279331.

  41. Jimenez Luna J, Grisoni F, Weskamp N, Schneider G. Artificial intelligence in drug discovery: recent advances and future perspectives. Expert Opin Drug Discov. 2021;16(9):949-59. doi: 10.1080/17460441.2021.1909567, PMID 33779453.

  42. Mak KK, Pichika MR. Artificial intelligence in drug development: present status and future prospects. Drug Discov Today. 2019;24(3):773-80. doi: 10.1016/j.drudis.2018.11.014, PMID 30472429.

  43. Reker D, Barbosa AJ. Navigating chemical space by interfacing generative artificial intelligence and molecular docking. J Chem Inf Model. 2020;60(3):1203-10.

  44. Cherkasov A, Muratov EN, Fourches D, Varnek A, Baskin II, Cronin M. QSAR modeling: where have you been? Where are you going to? J Med Chem. 2014;57(12):4977-5010. doi: 10.1021/jm4004285, PMID 24351051.

  45. Filipp FV. Opportunities for artificial intelligence in advancing precision medicine. Curr Genet Med Rep. 2019;7(4):208-13. doi: 10.1007/s40142-019-00177-4, PMID 31871830.

  46. Aliper A, Plis S, Artemov A, Ulloa A, Mamoshina P, Zhavoronkov A. Deep learning applications for predicting pharmacological properties of drugs and drug repurposing using transcriptomic data. Mol Pharm. 2016;13(7):2524-30. doi: 10.1021/acs.molpharmaceut.6b00248, PMID 27200455.

  47. Segler MH, Preuss M, Waller MP. Planning chemical syntheses with deep neural networks and symbolic AI. Nature. 2018;555(7698):604-10. doi: 10.1038/nature25978, PMID 29595767.

  48. Yang X, Wang Y, Byrne R, Schneider G, Yang S. Concepts of artificial intelligence for computer-assisted drug discovery. Chem Rev. 2019;119(18):10520-94. doi: 10.1021/acs.chemrev.8b00728, PMID 31294972.

  49. Tran PT, Bender A, Ho TT. Recent advances in artificial intelligence applications for drug discovery. Curr Opin Struct Biol. 2022;72:29-38.

  50. Gayvert KM, Madhukar NS, Elemento O. A data-driven approach to predicting successes and failures of clinical trials. Cell Chem Biol. 2016;23(10):1294-301. doi: 10.1016/j.chembiol.2016.07.023, PMID 27642066.

  51. Poongodi T, Lucia Agnesbeena TL, Janarthanan S, Balusamy B. Accelerating data acquisition process in the pharmaceutical industry using internet of things. An Industrial IoT Approach for Pharmaceutical Industry Growth. Amsterdam: Elsevier; 2020. p. 117-52. doi: 10.1016/B978-0-12-821326-1.00005-X.

  52. Singh M, Sachan S, Singh A, Singh KK. Internet of Things in pharma industry: possibilities and challenges. In: Emergence of pharmaceutical industry growth with industrial IoT approach. Amsterdam: Elsevier; 2020. p. 195-216. doi: 10.1016/B978-0-12-819593-2.00007-8.

  53. Alagarsamy S, Kandasamy R, Subbiah L, Palanisamy S. Applications of internet of things in pharmaceutical industry. SSRN Journal. 2019 Aug 23:14. doi: 10.2139/ssrn.3441099.

  54. Sharma A, Kaur J, Singh I. Internet of things (IoT) in pharmaceutical manufacturing warehousing and supply chain management. SN Comput Sci. 2020;1(4):212.

  55. Mishra S, Dash A, Mishra BK. An insight of internet of things applications in pharmaceutical domain. In: Emergence of pharmaceutical industry growth with industrial IoT approach. Elsevier; 2020. p. 245-73. doi: 10.1016/B978-0-12-819593-2.00009-1.

  56. Ogbuagu OO, Mbata AO, Balogun OD, Oladapo O, Ojo OO, Muonde M. Quality assurance in pharmaceutical manufacturing: bridging the gap between regulations supply chain and innovations. IJMRGE. 2023;4(1):823-31. doi: 10.54660/.IJMRGE.2023.4.1-823-831.

  57. Kodumuru R, Sarkar S, Parepally V, Chandarana J. Artificial intelligence and internet of things integration in pharmaceutical manufacturing: a smart synergy. Pharmaceutics. 2025;17(3):290. doi: 10.3390/pharmaceutics17030290, PMID 40142954.

  58. Rakshit P, Srivastava PK. Cutting edge IoT technology for smart Indian pharma. In: Int Conf Adv Comput Innov Technol Eng (ICACITE). New York: IEEE; 2021. doi: 10.1109/ICACITE51222.2021.9404627.

  59. Sharma DK, Gupta P, Priety. Internet of things: the new Rx for pharmaceutical manufacturing and supply chains. In: an industrial IoT approach for pharmaceutical industry growth. Amsterdam: Elsevier; 2020. p. 257-88. doi: 10.1016/B978-0-12-821326-1.00010-3.

  60. Shakhatreh M. Safeguarding pharmaceutical shipments using internet of things. Internet Things. 2020;12:100300.

  61. Ng EH, Nepal B, Schott E, Keathley H. Internet of things: opportunities and applications in pharmaceutical manufacturing and logistics. Proceedings of the am Soc Eng Manage. 2018;5(1):1-10.

  62. De Silva PY, Samarasinghe G. The impact of the internet of things on the efficiency of healthcare service delivery: a systematic review. J Med Internet Res. 2022;24(11):e39641.

  63. Ding B. Pharma industry 4.0: literature review and research opportunities in sustainable pharmaceutical supply chains. Process Saf Environ Prot. 2018 Oct;119:115-30. doi: 10.1016/j.psep.2018.06.031.

  64. Kaitin KI. Deconstructing the drug development process: the new face of innovation. Clin Pharmacol Ther. 2010;87(3):356-61. doi: 10.1038/clpt.2009.293, PMID 20130565.

  65. Bader F, Rahimifard S. Challenges for industrial robot applications in food manufacturing. In: proceedings of the 2nd international symposium on computer science and intelligent control. New York, USA: ACM; 2018. p. 1-8. doi: 10.1145/3284557.3284723.

  66. Calabrese GS, Pissavini S. From batch to continuous flow processing in chemicals manufacturing. AIChE J. 2011;57(4):828-34. doi: 10.1002/aic.12598.

  67. Jagtap S, Bhatt C, Thik J, Rahimifard S. Monitoring potato waste in food manufacturing using image processing and internet of things approach. Sustainability. 2019;11(11):3173. doi: 10.3390/su11113173.

  68. Lee J, Bagheri B, Kao HA. A cyber-physical systems architecture for industry 4.0-based manufacturing systems. Manuf Lett. 2015 Jan;3:18-23. doi: 10.1016/j.mfglet.2014.12.001.

  69. Lee WS, Grosh DL, Tillman FA, Lie CH. Fault tree analysis methods and applications: a review. IEEE Trans Reliab. 1985;34(3):194-203. doi: 10.1109/TR.1985.5222114.

  70. Jalal Ali HS, Hamdan A. The impact of blockchain in tracking the pharmaceuticals. Contrib Manag Sci. 2023 Oct 29:757-67. doi: 10.1007/978-981-99-6101-6_56.

  71. Mudasir Malla A, Banka AA. Pharma blocks. In: blockchain-based internet of things. Boca Raton: Chapman and Hall/CRC; 2024. p. 87-108. doi: 10.1201/9781003407096-6.

  72. Sahoo S. Blockchain-enabled traceability systems for supply chain quality management: empirical insights from pharmaceutical manufacturers. Int J Qual Reliab Manag. 2025;42(5):1381-410. doi: 10.1108/IJQRM-03-2024-0091.

  73. Cai X, Xu X, Li Q. Pharmaceutical cold chain logistics traceability system based on blockchain technology. In: Int Conf Distrib Comput Electr Circuits Electron (ICDCECE). New York: IEEE; 2023. p. 1-6. doi: 10.1109/ICDCECE57866.2023.10150534.

  74. Asha K, Anil George KA. Reducing counterfeit medicine through blockchain. In: Blockchain technology for IoE. New York: CRC Press; 2023. p. 239-59. doi: 10.1201/9781003366010-14.

  75. Akram W, Joshi R, Haider T, Sharma P, Jain V, Garud N. Blockchain technology: a potential tool for the management of pharma supply chain. Res Social Adm Pharm. 2024;20(6):156-64. doi: 10.1016/j.sapharm.2024.02.014, PMID 38423927.

  76. Ahmad RW, Al Khader W, Jayaraman R, Salah K, Antony J, Swarnakar V. Integrating lean six sigma with blockchain technology for quality management a scoping review of current trends and future prospects. TQM J. 2023;35(7):1609-31. doi: 10.1108/TQM-06-2022-0181.

  77. Ullagaddi P. Digital transformation in the pharmaceutical industry: enhancing quality management systems and regulatory compliance. Int J Health Sci. 2024;12(1):31-43. doi: 10.15640/ijhs.v12n1a4.

  78. Yik MH, Wong VC, Wong TH, Shaw PC. Herbchain, a blockchain based informative platform for quality assurance and quality control of herbal products. J Tradit Complement Med. 2021;11(6):598-600. doi: 10.1016/j.jtcme.2021.07.005, PMID 34765524.

  79. Haq I, Muselemu O. Blockchain technology in pharmaceutical industry to prevent counterfeit drugs. IJCA. 2018;180(25):8-12. doi: 10.5120/ijca2018916579.

  80. Tseng JH, Liao YC, Chong B, Liao SW. Governance on the drug supply chain via Gcoin blockchain. Int J Environ Res Public Health. 2018;15(6):1055. doi: 10.3390/ijerph15061055, PMID 29882861.

  81. Casino F, Dasaklis TK, Patsakis C. A systematic literature review of blockchain-based applications: current status, classification and open issues. Telemat Inform. 2019;36:55-81. doi: 10.1016/j.tele.2018.11.006.

  82. Sharma A, Kumar R, Chavali P. A framework for the application of blockchain in pharmaceutical supply chain. J Glob Oper Strateg Sourc. 2022;15(3):412-35.

  83. Seyedghorban Z, Tahernejad H, Meriton R, Graham G. Supply chain digitalization: past, present and future. Prod Plan Control. 2020;31(2-3):96-114. doi: 10.1080/09537287.2019.1631461.

  84. Korpela K, Hallikas J, Dahlberg T. Digital supply chain transformation toward blockchain integration. Proceedings of the Annual Hawaii International Conference on System Sciences. 2017:4182-91. doi: 10.24251/HICSS.2017.506.

  85. Beck R, Muller Bloch C, King JL. Governance in the blockchain economy: a framework and research agenda. J Assoc Inf Syst. 2018;19(10):1020-34. doi: 10.17705/1jais.00518.

  86. Chang SE, Chen YC, Wu TC. Exploring blockchain technology in international trade: business process re-engineering for letter of credit. Ind Manag Data Syst. 2019;119(8):1712-33. doi: 10.1108/IMDS-12-2018-0568.

  87. Zhang Y, Kasahara S, Shen Y, Jiang X, Wan J. Smart contract-based access control for the internet of things. IEEE Internet Things J. 2019;6(2):1594-605. doi: 10.1109/JIOT.2018.2847705.

  88. Salah K, Nizamuddin N, Jayaraman R, Omar M. Blockchain based soybean traceability in agricultural supply chain. IEEE Access. 2019;7:73295-305. doi: 10.1109/ACCESS.2019.2918000.

  89. Pal K, Yasar AU. Internet of things and blockchain technology in apparel manufacturing supply chain data management. Procedia Comput Sci. 2020;170:450-7. doi: 10.1016/j.procs.2020.03.088.

  90. Hsu CC, Sandford BA. The delphi technique: making sense of consensus. Pract Assess Res Eval. 2007;12(10):1-8. doi: 10.7275/pdz9-th90.

  91. Liao DY, Wang X. Applications of blockchain technology to logistics management in integrated casinos and entertainment. Informatics. 2018;5(4):44. doi: 10.3390/informatics5040044.

  92. Li Z, Kang J, Yu R, Ye D, Deng Q, Zhang Y. Consortium blockchain for secure energy trading in industrial internet of things. IEEE Trans Ind Inf. 2017;14(8):3690-700. doi: 10.1109/TII.2017.2786307.

  93. World Health Organization. WHO global surveillance and monitoring system for substandard and falsified medical products. Geneva: World Health Organization; 2017.

  94. Alzahrani N, Bulusu N. Block supply chain: a new anti-counterfeiting supply chain using NFC and blockchain. In: Proceedings of the 1st workshop on cryptocurrencies and blockchains for distributed systems. New York, USA: ACM; 2018. p. 30-5. doi: 10.1145/3211933.3211939.

  95. Chakraborty S, Aich S, Kim HC. A secure healthcare system design framework using blockchain technology. In: 21st International Conference on Advanced Communication Technology (ICACT). IEEE; 2019. p. 260-4. doi: 10.23919/ICACT.2019.8701983.

  96. Jamil F, Iqbal N, Imran S, Ahmad S, Kim D. Peer-to-peer energy trading mechanism based on blockchain and machine learning for sustainable electrical power supply in smart grid. IEEE Access. 2021;9:39193-217. doi: 10.1109/ACCESS.2021.3060457.

  97. Azzi R, Chamoun RK, Sokhn M. The power of a blockchain-based supply chain. Comput Ind Eng. 2019;135:582-92. doi: 10.1016/j.cie.2019.06.042.

  98. Gualandris J, Klassen RD, Vachon S, Kalchschmidt M. Sustainable evaluation and verification in supply chains: aligning and leveraging accountability to stakeholders. J Oper Manag. 2015;38(1):1-13. doi: 10.1016/j.jom.2015.06.002.

  99. Kamath R. Food traceability on blockchain: WalMart’s pork and mango pilots with IBM. The JBBA. 2018;1(1):1-12. doi: 10.31585/jbba-1-1-(10)2018.

  100. Angraal S, Krumholz HM, Schulz WL. Blockchain technology: applications in health care. Circ Cardiovasc Qual Outcomes. 2017;10(9):e003800. doi: 10.1161/CIRCOUTCOMES.117.003800, PMID 28912202.

  101. Clauson KA, Breeden EA, Davidson C, Mackey TK. Leveraging blockchain technology to enhance supply chain management in healthcare. Blockchain Healthc Today. 2018;1:1-12. doi: 10.30953/bhty.v1.20.

  102. Jamil F, Ahmad S, Iqbal N, Kim DH. Towards a remote monitoring of patient vital signs based on IoT-based blockchain integrity management platforms in smart hospitals. Sensors (Basel). 2020;20(8):2195. doi: 10.3390/s20082195, PMID 32294989.

  103. Kumar A, Liu R, Shan Z. Is blockchain a silver bullet for supply chain management? Technical challenges and research opportunities. Decis Sci. 2020;51(1):8-37. doi: 10.1111/deci.12396.

  104. Lacity MC. Addressing key challenges to making enterprise blockchain applications a reality. MIS Q Exec. 2018;17(3):201-22.

  105. Zheng Z, Xie S, Dai HN, Chen X, Wang H. Blockchain challenges and opportunities: a survey. Int J Web Grid Serv. 2018;14(4):352-75. doi: 10.1504/IJWGS.2018.095647.

  106. Yeung K. Regulation by blockchain: the emerging battle for supremacy between the code of law and code as law. Mod Law Rev. 2019;82(2):207-39. doi: 10.1111/1468-2230.12399.

  107. Tonnissen S, Teuteberg F. Analysing the impact of blockchain technology for operations and supply chain management: an explanatory model drawn from multiple case studies. Int J Inf Manag. 2020;52:101953. doi: 10.1016/j.ijinfomgt.2019.05.009.

  108. Carson B, Romanelli G, Walsh P, Zhumaev A. Blockchain beyond the hype: what is the strategic business value? McKinsey and Company; 2018.

  109. Jennings S, Ha K, Both D. Cetus 840®: a new paradigm for a modernized fully automated dissolution system. J Pharm Innov. 2009;4(3):107-14. doi: 10.1007/s12247-009-9062-6.

  110. Keller SH, Svarer C, Sibomana M. Attenuation correction for the HRRT PET-scanner using transmission scatter correction and total variation regularization. IEEE Trans Med Imaging. 2013;32(9):1611-21. doi: 10.1109/TMI.2013.2261313, PMID 23661313.

  111. Theanponkrang S, Suginta W, Weingart H, Winterhalter M, Schulte A. Robotic voltammetry with carbon nanotube-based sensors: a superb blend for convenient high-quality antimicrobial trace analysis. Int J Nanomedicine. 2015;10:859-68. doi: 10.2147/IJN.S75237, PMID 25670899.

  112. Brown AS, Badrick T. The next wave of innovation in laboratory automation: systems for auto-verification, quality control and specimen quality assurance. Clin Chem Lab Med. 2023;61(1):37-43. doi: 10.1515/cclm-2022-0409, PMID 36282956.

  113. Coito T, Martins MS, Firme B, Figueiredo J, Vieira SM, Sousa JM. Assessing the impact of automation in pharmaceutical quality control labs using a digital twin. J Manuf Syst. 2022;62:270-85. doi: 10.1016/j.jmsy.2021.11.014.

  114. Nunavath RS, Nagappan K. Future of pharmaceutical industry: role of artificial intelligence automation and robotics. J Pharmacol Pharmacother. 2024;15(2):85-94.

  115. Han Y, Makarova E, Ringel M, Telpis V. Digitization automation and online testing: the future of pharma quality control [McKinsey and Company report]; 2019.

  116. Alahmri MN, Alshehri AM, Gazwani HA, Alsemari SA. The role of automation in modern clinical laboratories. Tec Empresarial. 2024;6(2):762-73.

  117. Kamaraj A, Puranik S. Automation in pharmaceutical industry and global regulatory compliance. J Curr Pharm Res. 2023;17(4):13-25.

  118. Koido K, Watanabe D, Akiyama Y, Fukui K. A fully automated dissolution testing system with online HPLC analysis for dissolution studies of controlled release tablets. Pharmazie. 2022;77(9):350-6.

  119. Kumar S, Paruchuri S, Ketkar A, Kumar A. Facilitating clinical laboratory automation through robotics and artificial intelligence. J Lab Med. 2022;46(1):11-23.

  120. Kuila D, Bandyopadhyay D, Layek M, Besra K. Automating pharmaceutical quality control and assurance processes: benefits and challenges. Int J Pharm Sci Rev Res. 2023;78(2):135-42.

  121. Grebner C, Malmerberg E, Shewmaker A, Batista J, Nicholls A, Sadowski J. Virtual screening in the cloud: how big is big enough? J Chem Inf Model. 2020;60(9):4274-82. doi: 10.1021/acs.jcim.9b00779, PMID 31682421.

  122. Barkley WE, Callas D, Doshi S, Hyman R. Pharmaceutical laboratory automation adoption: current trends and future considerations. J Autom Methods Manag Chem. 2022;4(1):23-31.

  123. Narayana M, Lakshmana RS, Timmayya S, Rajesh R. Digital transformation in pharmaceutical quality control: from manual to automated workflows. J Pharm Anal. 2023;13(4):301-11.

  124. Schneider G. Automating drug discovery. Nat Rev Drug Discov. 2018;17(2):97-113. doi: 10.1038/nrd.2017.232, PMID 29242609.

  125. Bhagwat AM, Goswami S, Patel S, Prasad G. Impact of laboratory automation on pharmaceutical quality control: a meta-analysis of productivity and error rates. J Lab Autom. 2023;28(6):482-91.

  126. Brindle E, Nishizono T, Masci P, Rae C, Butler R, Sohoni P. Automation of analytical methods for pharmaceutical quality control and assurance. J Flow Chem. 2021;11(2):113-27.

  127. Patel RD, Sharma KH, Mehta TR, Lakshminarayana D. Statistical analysis of measurement uncertainty in pharmaceutical analysis: impact of automation on analytical variability. J Pharm Biomed Anal. 2023;218:114973.

  128. Rooney TM, Erickson JA, Hahn K, Joshi V, Hoener B, Havel J. Development and validation of an automated microsampling LC-MS/MS method for the quantitation of ledipasvir in human plasma. Bioanalysis. 2017;9(14):1067-78.

  129. Mullen M, Dieguez G, Liang M, Graham JB. Laboratory automation and digital transformation: impact on contract research organization laboratory efficiency. Pharm Technol. 2022;46(11):22-8.

  130. DiMasi JA, Grabowski HG, Hansen RW. Innovation in the pharmaceutical industry: new estimates of R & D costs. J Health Econ. 2016;47:20-33. doi: 10.1016/j.jhealeco.2016.01.012, PMID 26928437.

  131. Abuhelwa AY, Foster DJ, Mudge S, Hayes D, Upton RN. Population pharmacokinetic modeling of itraconazole and hydroxyitraconazole for oral SUBA-itraconazole and sporanox capsule formulations in healthy subjects in fed and fasted states. Antimicrob Agents Chemother. 2015;59(9):5681-96. doi: 10.1128/AAC.00973-15, PMID 26149987.

  132. Alomar MJ. Factors affecting the development of adverse drug reactions (Review article). Saudi Pharm J. 2014;22(2):83-94. doi: 10.1016/j.jsps.2013.02.003, PMID 24648818.

  133. Allison JJ, Wall TC, Spettell CM, Calhoun J, Fargason CA, Kobylinski RW. The art and science of chart review. Jt Comm J Qual Improv. 2000;26(3):115-36. doi: 10.1016/s1070-3241(00)26009-4, PMID 10709146.

  134. Bateman DN, Carroll R, Pettie J, Yamamoto T, Elamin ME, Peart L. Effect of the UK’s revised paracetamol poisoning management guidelines on admissions, adverse reactions and costs of treatment. Br J Clin Pharmacol. 2014;78(3):610-8. doi: 10.1111/bcp.12362, PMID 24666324.

  135. Cho I, Park H, Choi YJ, Hwang MH, Bates DW. Understanding the nature of medication errors in an ICU with a computerized physician order entry system. PLOS One. 2014;9(12):e114243. doi: 10.1371/journal.pone.0114243, PMID 25526059.

  136. Ahmed SP, Unnikrishnan MK, Sharma N. Pharmacoeconomics in Indian pharmaceutical education system: challenges and opportunities. Indian J Pharm Educ Res. 2022;56(3):686-92.

  137. McDowall RD. Validation of computerized analytical and networked systems. J Pharm Biomed Anal. 2023;26(3):507-15.

  138. Bhamra SK, Desai A, Imani Asrai R, Wolfram P. Blockchain in pharmaceutical supply chain: current applications, challenges and opportunities. Int J Pharm. 2022;621:121727.

  139. Chiarello RJ, McMahon WW. Delegated prescription privileges: expanded opportunities for the profession of pharmacy. Soc Sci Med. 2007;36(11):1491-8.

  140. Davis T, Hounkpe Ahissou Y, Mensah KA, Sanvee GM. Current status of laboratory automation in pharmaceutical industry: a systematic review. J Lab Autom. 2022;27(5):391-406.

  141. Dougherty AP, Young S, Suen M, Grosser ST. Laboratory automation strategies: A survey of pharmaceutical quality control laboratory directors. Pharm Technol. 2023;47(1):36-43.