View Article

Abstract

The pharmaceutical industry is undergoing a paradigm shift, driven by the rapid advancement of Artificial Intelligence (AI) and Machine Learning (ML) technologies. Traditionally reliant on retrospective analyses, manual inspection, and standardized quality control protocols, pharmaceutical quality assurance (QA) is evolving into a dynamic, data-driven discipline. This transformation is largely due to the integration of AI and ML, which enable real-time monitoring, predictive analytics, and intelligent decision making across the product lifecycle. This review explores the foundational concepts of AI and ML, their specific applications in pharmaceutical QA, and the benefits these technologies offer in enhancing process control, product consistency, and regulatory compliance. Applications such as predictive maintenance, real-time release testing (RTRT), automated visual inspection, and natural language processing (NLP) for documentation review demonstrate significant improvements in operational efficiency, cost-effectiveness, and risk mitigation. The paper also highlights the adoption of AI-driven systems by leading pharmaceutical companies including Pfizer, GSK, and Sanofi, who have successfully implemented ML algorithms for process optimization and stability prediction. Furthermore, the review discusses current regulatory guidelines from bodies such as the US FDA, EMA, and ICH, which are beginning to address the use of AI/ML in regulated environments through frameworks like Good Machine Learning Practice (GMLP). Despite the immense potential, the implementation of AI and ML in QA is not without challenges. Issues related to data quality, model validation, regulatory acceptance, and integration with legacy systems remain critical barriers. In addition, ethical considerations regarding algorithm transparency, bias, and data privacy must be addressed for wider acceptance. Looking ahead, the emergence of Explainable AI (XAI), digital twins, federated learning, and blockchain integration point toward a more transparent, secure, and intelligent QA ecosystem. The successful realization of these advancements will require a collaborative effort among industry stakeholders, regulatory agencies, and academic researchers to establish standards, ensure compliance, and build trust in AI-enabled quality systems.

Keywords

Artificial Intelligence, Quality Assurance, Machine Learning, Automated Visual Inspection, Validation, Data Integrity.

Introduction

The pharmaceutical industry is one of the most regulated and quality-sensitive sectors in the world, where product integrity, patient safety, and regulatory compliance are paramount. Over the past decades, pharmaceutical quality assurance (QA) has evolved from simple inspection-based activities to a more holistic approach that ensures quality is built into every stage of the drug lifecycle—from raw material procurement to product release.

However, traditional QA systems are increasingly challenged by the growing complexity of drug formulations, globalized manufacturing networks, and the rising demand for faster production cycles. These systems often depend on retrospective quality control, periodic audits, and rigid standard operating procedures, which may not fully address modern manufacturing dynamics. As a result, there is a compelling need for smarter, adaptive, and data-driven QA mechanisms.

Artificial Intelligence (AI) and (ML) have emerged as powerful tools to bridge this gap. AI refers to computational systems capable of mimicking human intelligence, while ML, a subset of AI, involves algorithms that learn patterns from data and improve over time without being explicitly programmed. These technologies have already demonstrated success in domains such as drug discovery, clinical trial design, and supply chain optimization. Their integration into QA practices promises enhanced efficiency, reduced human error, and proactive risk mitigation.

With Pharma 4.0 and Industry 4.0 driving digital transformation across pharmaceutical operations, AI and ML are being deployed for predictive maintenance, real-time process monitoring, automated deviation analysis, and regulatory compliance assurance. Moreover, advanced AI tools such as deep learning and natural language processing (NLP) are transforming document review processes and visual inspection techniques.

This paper aims to provide a comprehensive review of the role and impact of AI and ML in pharmaceutical quality assurance. It outlines their core principles, real-world applications, regulatory considerations, challenges in adoption, and future trends. By consolidating the current knowledge and industrial insights, this work contributes to a better understanding of how AI and ML can reshape the future of pharmaceutical QA.

2. Evolution of AI and ML in the Pharmaceutical Industry:

The application of Artificial Intelligence (AI) and Machine Learning (ML) in the pharmaceutical sector has grown from niche experimental tools to integral components of drug development and quality systems. The evolution of these technologies has paralleled advancements in computational power, data storage capabilities, and algorithmic sophistication.

2.1. Early Beginnings:

The roots of AI in healthcare and pharmaceuticals can be traced back to the 1970s and 1980s, when rule-based expert systems like MYCIN and DENDRAL were developed to support clinical decision-making and chemical structure elucidation. However, these systems were limited by their dependency on predefined rules and static logic.

In the 1990s and early 2000s, data mining and statistical learning techniques began to gain attention in bioinformatics and pharmacovigilance. At this stage, the focus remained largely on research-related applications, such as analyzing gene sequences, predicting drug-target interactions, and optimizing clinical trial recruitment.

2.2. The Rise of ML in Drug Discovery and Development:

The past two decades have seen a significant expansion in the use of machine learning, particularly in drug discovery, clinical trial design, and personalized medicine. Supervised and unsupervised learning models, along with neural networks, enabled the prediction of compound activity, toxicity, and patient outcomes with increasing accuracy. Tools such as Bayesian networks, support vector machines, and random forests were adopted for analyzing high-throughput screening data and patient health records.

2.3. Transition to Manufacturing and Quality Assurance:

More recently, the application of AI and ML has extended beyond research and development into the manufacturing and quality assurance domains, aligned with the digital transformation initiatives of Industry 4.0 and Pharma 4.0. This shift has been fueled by several factors:

  1. Availability of big data from manufacturing execution systems (MES), laboratory information management systems (LIMS), and process analytical technologies (PAT).
  2. Regulatory encouragement of innovation through Quality by Design (QbD) and Real-Time Release Testing (RTRT).
  3. The need for predictive quality systems that reduce human intervention and improve process robustness.

Pharmaceutical companies began implementing AI models for predictive maintenance, process parameter optimization, real-time quality monitoring, and automated deviation detection. AI-powered computer vision has been used to enhance visual inspections, while natural language processing (NLP) tools have automated the review of standard operating procedures (SOPs), batch manufacturing records (BMRs), and audit documentation.

2.4. The Present and Future:

Today, AI and ML are recognized as critical enablers of adaptive quality systems. The integration of these technologies is no longer optional but essential for companies aiming to remain competitive, compliant, and agile in an increasingly complex regulatory and market landscape.

Looking ahead, emerging trends such as explainable AI (XAI), federated learning, digital twins, and blockchain are expected to further transform pharmaceutical QA by increasing transparency, decentralization, and trustworthiness in automated decision-making systems.

3. Core Concepts and Terminologies in AI and ML for Pharmaceutical Quality Assurance:

To understand the role of Artificial Intelligence (AI) and Machine Learning (ML) in pharmaceutical quality assurance (QA), it is essential to define and differentiate the core concepts and terminologies that underlie these technologies. This section outlines key definitions and their relevance to pharmaceutical QA systems.

3.1 Artificial Intelligence (AI)

AI refers to the capability of computer systems to mimic human intelligence to perform tasks such as learning, reasoning, problem-solving, decision-making, and language understanding. In QA, AI enables systems to automate decision processes, identify deviations, and suggest corrective actions without human input.

Types of AI in pharma QA:

  1. Narrow AI: AI systems designed to perform specific tasks (e.g., image-based tablet inspection).
  2. General AI: Still theoretical in pharma, general AI refers to machines with generalized intelligence across multiple domains.

3.2 Machine Learning (ML)

ML is a subset of AI that uses statistical techniques to allow machines to improve with experience. ML algorithms learn patterns from historical data and make predictions or decisions based on new inputs. This is highly valuable in predicting process deviations or quality defects before they occur.

Common ML types used in QA:

  1. Supervised Learning: Algorithms learn from labeled data (e.g., defect or no defect).
  2. Unsupervised Learning: Algorithms find patterns in unlabeled data (e.g., clustering anomalies).
  3. Reinforcement Learning: Systems learn optimal actions through trial and error based on feedback (e.g., adaptive process control).

3.3 Deep Learning (DL)

A specialized form of ML based on neural networks with multiple layers. Deep learning is particularly powerful in recognizing complex patterns in large datasets, such as detecting defects in images of tablets or capsules. DL models are used in computer vision for visual inspection and in natural language processing for document analysis

3.4 Natural Language Processing (NLP)

NLP is a field within AI that enables machines to understand, interpret, and generate human language. In QA, NLP can be used to automate the review of SOPs, BMRs, audit trails, and regulatory documents, ensuring compliance and reducing human errors.

3.5 Computer Vision

Computer vision involves the use of AI to analyze visual inputs such as images or video frames. In pharmaceutical QA, it enables real-time visual inspection of tablets, vials, and packaging, identifying defects like cracks, discoloration, or label mismatches more accurately and faster than manual inspection.

3.6 Process Analytical Technology (PAT)

PAT is a system for designing, analyzing, and controlling pharmaceutical manufacturing through timely measurements of critical quality and performance attributes. ML can be integrated with PAT to enable continuous process monitoring and real-time decision making.

3.7 Quality by Design (QbD) and Real-Time Release Testing (RTRT)

QbD is a systematic approach to pharmaceutical development that emphasizes designing quality into processes from the outset. ML supports QbD by identifying critical process variables and optimizing them.

RTRT allows batch release based on real-time data rather than traditional end-product testing. AI-driven models can predict product quality during the manufacturing process, supporting RTRT adoption.

4. Applications of AI and ML in Pharmaceutical Quality Assurance:

The integration of AI and ML into pharmaceutical quality assurance (QA) systems is transforming traditional practices by improving efficiency, reducing human error, and enabling real-time, data-driven decisions. These technologies are applied across various stages of the product lifecycle, from manufacturing to release and postmarketing surveillance. Below are key application areas where AI and ML are making significant impacts:

4.1 Predictive Quality Analytics

AI and ML algorithms can analyze historical process and quality data to predict potential failures or deviations before they occur. These models identify correlations between input parameters and final product quality, allowing manufacturers to take proactive steps.

Example:

Predicting tablet hardness or dissolution rate based on process parameters like granulation time or compression force.

4.2 Real-Time Process Monitoring and Control

Using machine learning with Process Analytical Technology (PAT), real-time data from sensors can be used to monitor critical quality attributes (CQAs) during manufacturing. AI systems automatically adjust process parameters to maintain quality within specifications.

Benefits:

  • Enables Real-Time Release Testing (RTRT)
  • Reduces batch failure rates
  • Enhances process robustness

4.3 Automated Visual Inspection

Traditional visual inspection is time-consuming and prone to human error. AI-powered computer vision systems can inspect tablets, capsules, vials, and packaging in real time using high-resolution cameras and deep learning models.

Capabilities:

  • Detecting cracks, chips, and discoloration
  • Identifying label defects or misalignment
  • Sorting defective products with high accuracy

4.4 Natural Language Processing (NLP) for Documentation Review

Pharmaceutical QA involves extensive documentation review, including SOPs, Batch Manufacturing Records (BMRs), and audit reports. NLP tools can read, analyze, and summarize documents, flagging inconsistencies or missing entries.

Use Cases:

  • Automated compliance checks
  • Faster audit readiness
  • Intelligent document classification and retrieval.

4.5 Deviations and CAPA Management

AI tools can assist in root cause analysis (RCA) by identifying patterns in deviations, out of specification (OOS) events, and complaints. ML algorithms prioritize corrective and preventive actions (CAPA) based on historical effectiveness.

Outcomes:

  • Shorter investigation times
  • More effective corrective actions
  • Reduced recurrence of deviations

4.6 Predictive Maintenance of Equipment

ML models can predict equipment failures by analyzing sensor data such as temperature, vibration, and pressure. This allows for preventive maintenance before breakdowns occur, minimizing downtime and ensuring continuous production.

Impact:

  • Reduces unplanned shutdowns
  • Increases equipment life
  • Improves operational efficiency

4.7 Regulatory Compliance and Audit Preparation

AI solutions can ensure compliance by continuously monitoring data integrity, traceability, and adherence to 21 CFR Part 11, Annex 11, and ICH Q10 guidelines. NLP tools help prepare for audits by organizing documentation and extracting key information.

4.8 Digital Twin Models for QA

A digital twin is a virtual replica of a manufacturing process. AI-powered digital twins allow QA teams to simulate changes in process conditions and predict quality outcomes without altering actual production.

Advantages:

  • Enhances process understanding
  • Supports decision-making
  • Minimizes experimentation costs.

5. Case Studies and Industrial Implementation:

The adoption of AI and ML in pharmaceutical quality assurance is no longer limited to theoretical or experimental frameworks. Leading pharmaceutical companies have begun integrating these technologies into their QA systems, demonstrating measurable improvements in product quality, compliance, and process efficiency. This section presents selected real-world case studies that illustrate successful AI/ML applications across the industry.

5.1 Pfizer – Predictive Quality and Process Optimization

Pfizer has implemented machine learning models in its manufacturing operations to monitor and predict the performance of critical quality attributes. By analyzing large volumes of batch data, AI algorithms identify trends and deviations in real-time.

Use Case: ML was used to predict dissolution outcomes of oral solid dosage forms, leading to better control of granulation and compression parameters.

Outcome: Reduction in batch failures, improved process control, and faster product release decisions.

5.2 GSK – AI for Visual Inspection

GSK integrated computer vision systems powered by deep learning to enhance visual inspection of parenteral products. The system was trained using thousands of images of acceptable and defective units.

Use Case: Detection of particulate matter, label defects, and container damage during the fill-finish stage.

Outcome: Increased detection accuracy, reduced operator fatigue, and enhanced batch quality compliance.

5.3 Sanofi – Digital Twins and Real-Time Release

Sanofi has developed digital twin models to simulate and optimize manufacturing processes in real time. These AI-driven models replicate the behavior of actual equipment and processes, allowing QA teams to predict deviations.

Use Case: A digital twin of a bioreactor system was used to simulate upstream production and predict product yield and quality.

Outcome: Improved yield consistency and faster implementation of RTRT approaches.

5.4 Novartis – NLP for Document Automation

Novartis deployed Natural Language Processing (NLP) tools to automate the review of standard operating procedures (SOPs) and other quality documents.

Use Case: NLP was used to extract critical control points and identify non-compliance in batch manufacturing records.

Outcome: Shorter documentation review time, increased regulatory compliance, and faster audit response.

5.5 Merck – AI in Predictive Maintenance

Merck uses AI for predictive maintenance of manufacturing equipment, analyzing sensor data (e.g., vibration, temperature, acoustic signals) to forecast mechanical failures.

Use Case: Monitoring of tablet compression machines and HVAC systems.

Outcome: Reduced downtime, fewer production interruptions, and improved product consistency.

5.6 AstraZeneca – End-to-End AI Integration

AstraZeneca is investing heavily in end-to-end AI platforms that integrate drug development, manufacturing, and quality control systems. The company uses AI to unify data from multiple sources for a centralized QA dashboard.

Use Case: Integration of LIMS, MES, and PAT data for a comprehensive quality risk management platform.

Outcome: Enhanced traceability, centralized monitoring, and rapid decision-making.

Key Takeaways from Industry Implementation:

  1. Customization is key: Each AI/ML system must be tailored to the specific process and compliance requirements of the facility.
  2. Data quality determines model success: Clean, high-resolution, and well annotated data are essential for accurate predictions.
  3. Change management is crucial: Training, digital literacy, and organizational readiness are critical to adopting AI in regulated environments.
  4. Validation and regulatory transparency are essential for AI deployment in GMP settings.

6. Regulatory Perspective and Compliance Challenges:

The integration of Artificial Intelligence (AI) and Machine Learning (ML) into pharmaceutical Quality Assurance (QA) introduces both immense potential and significant regulatory challenges. As the pharmaceutical industry is highly regulated by global authorities such as the U.S. FDA, EMA, and WHO, the deployment of AI tools must align with existing regulatory frameworks while addressing concerns related to data integrity, validation, and accountability.

6.1 Regulatory Guidance and Initiatives

  • U.S. FDA (Food and Drug Administration):
      • Issued a discussion paper on the use of AI/ML in Software as a Medical Device (SaMD).
      • Promotes the use of Quality by Design (QbD) and Real-Time Release Testing (RTRT)—both of which can benefit from AI integration.
      • In 2023, the FDA launched an AI Regulatory Framework Initiative to assess AI applications in manufacturing and quality.
  • EMA (European Medicines Agency):
  • Emphasizes the use of data-rich tools and real-time monitoring under ICH Q10.
  • Supports digital transformation under Annex 11 for computerized systems, which can include AI-based platforms.
  • ICH Guidelines (International Council for Harmonization):
  • Guidelines like ICH Q8 (R2), Q9, and Q10 form the foundation for science- and risk-based approaches to quality, indirectly supporting AI-enabled risk management.

6.2 Key Compliance Considerations for AI/ML Systems

To ensure that AI-based systems are accepted by regulators, several compliance and validation steps must be addressed:

6.2.1 Validation and Verification of AI Models

  • Model Validation: AI models must be validated for their intended use, just like analytical methods or manufacturing equipment.
  • Model Explain ability: Regulatory bodies may reject black-box models if their decision-making logic cannot be explained (hence, Explainable AI (XAI) is preferred).
  • Revalidation: Continuous learning models need revalidation whenever significant updates occur.

6.2.2 Data Integrity (ALCOA+)

  • AI systems must adhere to ALCOA+ principles (Attributable, Legible, Contemporaneous, Original, Accurate, Complete, Consistent, Enduring, and Available).
  • Audit trails must be implemented to trace every input, model prediction, and decision.

6.2.3 GxP Compliance

  • AI solutions used in QA must comply with GxP (Good Automated Manufacturing Practice).
  • All software must be validated according to GAMP 5 guidelines.
  • AI must be integrated with risk-based approaches for implementation in GMP facilities.

6.2.4 Change Control and Lifecycle Management

  • All AI model changes must go through documented change control.
  • Lifecycle documents should include model training records, testing outcomes, performance metrics, and update logs.

6.2.5 Cybersecurity and Data Protection

  • AI systems must protect sensitive production and patient-related data.
  • Must comply with global data privacy laws (e.g., GDPR, HIPAA) in case of clinical or post-market QA surveillance.

6.3 Challenges in Regulatory Acceptance

Despite promising performance, AI integration into QA faces regulatory hurdles:

  • Lack of standardized frameworks for AI validation in GMP environments.
  • Difficulty in interpreting complex ML outputs for regulatory review.
  • Regulatory hesitation to approve adaptive AI systems that continuously evolve.
  • Concerns over bias in training datasets affecting decision fairness.

6.4 Collaborative and Future-Oriented Initiatives

Global regulatory bodies are collaborating with industry leaders to shape the future of AI in pharma QA:

  • PIC/S and ISPE are working on AI guidance specific to manufacturing.
  • FDA’s CDRH and CDER are piloting AI in automated inspections and review systems.
  • Industry groups are drafting AI Risk Management Frameworks to guide regulatory friendly AI development.

In conclusion, while regulatory frameworks for AI and ML in pharmaceutical QA are still evolving, existing guidelines provide a foundation for risk-based, validated, and transparent implementation. To gain regulatory trust, companies must ensure accountability, traceability, and explain ability in all AI-driven QA activities.

7. Challenges and Limitations in Adoption of AI and ML in Pharmaceutical Quality Assurance:

Despite the substantial benefits offered by Artificial Intelligence (AI) and Machine Learning (ML) in pharmaceutical quality assurance, their widespread adoption faces several critical barriers. These challenges are technological, regulatory, organizational, and ethical in nature, and understanding them is essential for successful integration and scaling within the pharmaceutical ecosystem.

7.1 Data-Related Challenges

7.1.1 Data Quality and Availability

AI and ML systems require large volumes of clean, structured, and high-resolution data. In pharmaceutical QA, such data may be:

  • Scattered across siloed systems (e.g., LIMS, MES, SCADA).
  • Incomplete or manually entered, leading to inconsistency.
  • Unstructured (e.g., handwritten batch records or PDFs).

Impact: Poor-quality data directly compromises model training, accuracy, and generalizability.

7.1.2 Data Privacy and Security

Pharmaceutical QA often deals with confidential process data and patient-related postmarketing data. Ensuring compliance with HIPAA, GDPR, and company-specific IT policies is critical when training AI systems.

7.2 Technological Barriers

7.2.1 Model Transparency and Explain ability

Many ML algorithms, especially deep learning models, function as "black boxes"—they make predictions without clear explanations. In a highly regulated QA environment, lack of explain ability hinders:

  • Regulatory acceptance
  • Root cause investigations
  • Trust from QA personnel

7.2.2 Integration with Legacy Systems

Most pharmaceutical manufacturing facilities use legacy software systems that are not compatible with AI platforms. Integration requires significant investment and may disrupt validated processes.

7.3 Regulatory and Validation Challenges

  • Uncertainty in Regulatory Expectations: Regulatory bodies are still developing detailed guidelines on AI/ML validation, especially for continuously learning models.
  • Model Validation and Lifecycle Management: Continuous retraining of AI models requires revalidation, version control, and robust documentation, increasing compliance complexity.
  • Audit Preparedness: Models must be able to justify decisions retrospectively— difficult for some AI systems without built-in traceability.

7.4 Human and Organizational Resistance

7.4.1 Lack of Digital Literacy

Many QA personnel are unfamiliar with AI/ML tools and may resist adoption due to:

  • Fear of job displacement
  • Inadequate training in digital tools
  • Misunderstanding of AI capabilities

7.4.2 Change Management

Implementing AI requires changes in workflows, risk assessment strategies, and cross functional collaboration. Organizations without a culture of digital innovation may face internal resistance.

7.5 Cost and Resource Constraints

  • High Initial Investment: Procuring AI software, data infrastructure, and skilled personnel can be costly.
  • Skilled Workforce Shortage: There is a scarcity of professionals with both domain expertise in pharmaceuticals and technical expertise in AI/ML.
  • Ongoing Maintenance: ML models require continuous data monitoring, model tuning, and validation efforts, increasing operational costs.

7.6 Ethical and Bias Concerns

  • Algorithmic Bias: AI models trained on biased datasets may make flawed predictions, especially in post-market surveillance or product defect analysis.
  • Decision Accountability: Who is responsible for errors made by AI—developers, QA managers, or software vendors?

 7.7 Limitations of AI Algorithms in QA

  • AI cannot yet fully replace human judgment in areas requiring contextual decision making, such as interpreting deviations.
  • AI struggles in low-volume, high-variation environments typical of R&D or small batch GMP manufacturing.

Challenge Area

Description

Data Quality & Security

Incomplete, siloed, and unstructured data hinder model training

Technology &

Integration

Difficulties in integrating AI with existing systems

Regulatory Compliance

Lack of clear guidelines and high validation burden

Workforce & Training

Low digital literacy and resistance to change

While these challenges are significant, they are not insurmountable. With strategic planning, cross-functional collaboration, and proactive engagement with regulators, AI and ML can be responsibly and effectively embedded into QA systems.

8.1. Perspectives and Emerging Trends:

The adoption of Artificial Intelligence (AI) and Machine Learning (ML) in pharmaceutical Quality Assurance (QA) is poised to evolve rapidly, driven by technological innovation, increasing data availability, and growing regulatory acceptance. Looking ahead, several emerging trends and future directions are set to redefine the landscape of QA in the pharmaceutical industry.

The traditional QA approach is often retrospective and reactive, where quality issues are detected post-manufacture. The future vision focuses on predictive quality assurance, enabled by:

  • Real-Time Analytics
  • Self-learning AI systems
  • Integrated Quality Risk Management (QRM) tools

Impact: Issues will be predicted and corrected before product release, aligning with Quality by Design (QbD) principles.

8.2 Explainable AI (XAI)

As regulatory bodies demand transparency in AI decision-making, Explainable AI will become essential. XAI tools aim to provide:

  • Human-understandable justifications for AI predictions
  • Visual mapping of model logic and decision pathways
  • Enhanced trust and compliance in regulated QA settings

Use Case: In batch failure investigations, XAI could explain which parameters contributed most to product rejection.

8.3 Federated Learning and Secure Data Sharing

Data sharing is essential for developing robust AI models, but pharma companies often operate in silos due to IP concerns. Federated learning offers a solution:

  • Allows multiple organizations to collaboratively train AI models without sharing        raw data
  • Preserves data privacy and complies with regulations
  • Enables cross-industry learning without compromising confidentiality

8.4 AI-Driven Quality Management Systems (QMS):

Next-generation QMS platforms will embed AI to automate:

  • CAPA recommendations
  • Risk prioritization
  • Document control and audit readiness

Example: An AI-QMS could flag trends in deviation reports and automatically initiate CAPA workflows.

8.5 Digital Twins for Quality Assurance

The use of digital twins—virtual replicas of physical processes—is expected to expand in QA:

  • Enable simulation of quality outcomes before actual batch runs
  • Optimize process parameters in silico
  • Reduce experimentation and raw material wastage

This will enhance the QbD framework and enable personalized process tuning.

8.6 AI-Powered Audits and Self-Inspections

In the future, AI-enabled internal audits could scan manufacturing and documentation systems to identify compliance gaps, out-of-specifications, or data integrity risks in real time.

  • Reduces audit preparation time
  • Enhances 24/7 compliance monitoring
  • Improves readiness for regulatory inspections

8.7 Blockchain Integration for Traceability

AI systems integrated with blockchain technology will enhance data transparency, traceability, and security, particularly in QA aspects like:

  • Electronic batch records (EBR)
  • Chain-of-custody for raw materials
  • Post-market surveillance data

This will fulfill both regulatory and public demands for **end-to-end product traceability.

8.8 Personalized QA Strategies Using AI

With the rise of precision medicine and small-batch biologics, AI will be used to develop personalized quality assurance protocols based on:

  • Product-specific risk profiles
  • Patient-specific stability factors
  • Dynamic process conditions

8.9 Global Regulatory Harmonization

Regulatory agencies (FDA, EMA, PMDA, CDSCO) are expected to collaborate globally to release unified AI validation and compliance guidelines.

AI-based quality tools may soon be part of common technical documents (CTDs)and GMP inspection checklists.

Future Outlook

Trend

Expected Benefit

Predictive QA Systems

Early detection and prevention of failures

Explainable AI

Regulatory trust and improved investigations

Digital Twins

Virtual process testing and QbD enhancement

AI-Embedded QMS

Automation of routine QA tasks

Blockchain-AI Integration

Full traceability and data integrity assurance

Federated Learning

Collaboration without data compromise

Regulatory Harmonization

Standardized AI compliance guidelines globally

The future of pharmaceutical quality assurance lies in proactive, intelligent, and autonomous systems that leverage the full power of AI and ML. As the industry CONTINUES to adopt digitalization, the next decade will likely witness a complete transformation in how pharmaceutical quality is monitored, controlled, and ensured.

CONCLUSION:

The integration of Artificial Intelligence (AI) and Machine Learning (ML) into pharmaceutical quality assurance (QA) represents a transformative shift in how the industry ensures the safety, efficacy, and consistency of medicinal products. These advanced technologies offer unparalleled capabilities in data analysis, process monitoring, deviation prediction, and automated decision-making, thereby reinforcing the foundational principles of Quality by Design (QbD) and Continuous Process Verification (CPV).

Throughout this review, it is evident that AI/ML applications in QA can significantly enhance operational efficiency, reduce human errors, improve compliance, and enable real-time, risk-based quality control. From predictive maintenance to digital twins, visual defect recognition to real-time release testing (RTRT), these technologies are reshaping conventional QA frameworks and aligning them with the goals of Industry 4.0 and Pharma 4.0.

However, their adoption is not without challenges. Issues related to data integrity, regulatory uncertainty, model explain ability, workforce readiness, and ethical accountability must be systematically addressed. Regulatory authorities like the FDA, EMA, and ICH are beginning to develop guidelines and frameworks that support innovation while upholding stringent compliance requirements. Future success depends on transparent validation practices, lifecycle management, and interdisciplinary collaboration between pharma professionals, data scientists, and regulatory experts.

Looking ahead, the evolution of Explainable AI (XAI), federated learning, block chain integration, and AI-augmented Quality Management Systems (QMS) will pave the way for more resilient, autonomous, and intelligent QA ecosystems. These advancements will not only streamline pharmaceutical manufacturing but also enhance public trust in medicine quality and patient safety.

In conclusion, while the road to full-scale AI and ML adoption in pharmaceutical QA may be complex, it is also inevitable. Companies that embrace these technologies responsibly and strategically will be better positioned to meet future regulatory expectations, achieve operational excellence, and deliver high-quality products in an increasingly data-driven healthcare environment.

REFERENCES

  1. STM International. (2020). ASTM E2500-20: Standard guide for specification, design, and verification of pharmaceutical and biopharmaceutical manufacturing systems and equipment. ASTM International. https://doi.org/10.1520/E2500-20
  2. ASTM International. (2017). ASTM E2968-17: Standard guide for application of continuous process verification to pharmaceutical and biopharmaceutical manufacturing. ASTM International.
  3. Bakeev, K. A. (Ed.). (2010). Process analytical technology: Spectroscopic tools and implementation strategies for the chemical and pharmaceutical industries (2nd ed.). Wiley. https://doi.org/10.1002/9780470689592
  4. Bishop, C. M. (2006). Pattern recognition and machine learning. Springer.
  5. Breiman, L. (2001). Random forests. Machine Learning, 45(1), 5–32. https://doi.org/10.1023/A:1010933404324
  6. Carvalho, T. P., Soares, F. A. A. M. N., Vita, R., Francisco, R. de P., Basto, J. P., & Alcalá, S. G. S. (2019). A systematic literature review of machine learning methods applied to predictive maintenance. Computers & Industrial Engineering, 137, 106024. https://doi.org/10.1016/j.cie.2019.106024
  7. Casino, F., Dasaklis, T. K., & Patsakis, C. (2019). A systematic literature review of blockchain-based applications: Current status, classification and open issues. Telematics and Informatics, 36, 55–81. https://doi.org/10.1016/j.tele.2018.11.006
  8. Cortes, C., & Vapnik, V. (1995). Support-vector networks. Machine Learning, 20(3), 273–297. https://doi.org/10.1007/BF00994018
  9. Devlin, J., Chang, M.-W., Lee, K., & Toutanova, K. (2019). BERT: Pre-training of deep bidirectional transformers for language understanding. In Proceedings of NAACL-HLT 2019 (pp. 4171–4186).
  10. Doshi-Velez, F., & Kim, B. (2017). Towards a rigorous science of interpretable machine learning. arXiv. https://arxiv.org/abs/1702.08608
  11. European Commission. (2022). EudraLex Volume 4: EU Guidelines for Good Manufacturing Practice for Medicinal Products for Human and Veterinary Use—Annex 1: Manufacture of sterile medicinal products (Revision).
  12. European Medicines Agency. (2011). Annex 11: Computerised systems (EU GMP, Volume 4).
  13. European Medicines Agency. (2012). Guideline on real time release testing (RTRT) of finished products. EMA/CHMP/QWP/811210/2009.
  14. Food and Drug Administration. (2004). Guidance for industry: PAT—A framework for innovative pharmaceutical development, manufacturing, and quality assurance. U.S. Department of Health and Human Services.
  15. Food and Drug Administration. (2011). Guidance for industry: Process validation—General principles and practices. U.S. Department of Health and Human Services.
  16. Food and Drug Administration. (2018). Data integrity and compliance with drug CGMP: Questions and answers—Guidance for industry. U.S. Department of Health and Human Services.
  17. Food and Drug Administration (CDER & CBER). (2023). Discussion paper: Using artificial intelligence & machine learning in the development of drug & biological products. U.S. Food and Drug Administration.
  18. Goodfellow, I., Bengio, Y., & Courville, A. (2016). Deep learning. MIT Press.
  19. Guidotti, R., Monreale, A., Ruggieri, S., Turini, F., Giannotti, F., & Pedreschi, D. (2018). A survey of methods for explaining black box models. ACM Computing Surveys, 51(5), 93. https://doi.org/10.1145/3236009
  20. ICH. (2008). ICH Q10: Pharmaceutical quality system. International Council for Harmonisation.
  21. ICH. (2009). ICH Q8(R2): Pharmaceutical development. International Council for Harmonisation.
  22. ICH. (2023). ICH Q9(R1): Quality risk management (Revision 1). International Council for Harmonisation.
  23. ICH. (2022). ICH Q13: Continuous manufacturing of drug substances and drug products. International Council for Harmonisation.
  24. ICH. (2023). ICH Q14: Analytical procedure development. International Council for Harmonisation.
  25. ISPE. (2017). GAMP® Guide: Records and data integrity. International Society for Pharmaceutical Engineering.
  26. ISPE. (2022). GAMP® 5, Second Edition: A risk-based approach to compliant GxP computerized systems. International Society for Pharmaceutical Engineering.
  27. Jardine, A. K. S., Lin, D., & Banjevic, D. (2006). A review on machinery diagnostics and prognostics implementing condition-based maintenance. Mechanical Systems and Signal Processing, 20(7), 1483–1510. https://doi.org/10.1016/j.ymssp.2005.09.012
  28. Jurafsky, D., & Martin, J. H. (2023). Speech and language processing (3rd ed., draft). Stanford University/Prentice Hall.
  29. Kone?ný, J., McMahan, H. B., Yu, F. X., Richtárik, P., Suresh, A. T., & Bacon, D. (2016). Federated learning: Strategies for improving communication efficiency. arXiv. https://arxiv.org/abs/1610.05492
  30. Kourti, E. (2005). Application of latent variable methods to process control and multivariate statistical process control in industry. International Journal of Adaptive Control and Signal Processing, 19(4), 213–246. https://doi.org/10.1002/acs.859
  31. Kuhn, M., & Johnson, K. (2013). Applied predictive modeling. Springer. https://doi.org/10.1007/978-1-4614-6849-3
  32. Lee, J., Bagheri, B., & Kao, H.-A. (2015). A cyber-physical systems architecture for industry 4.0-based manufacturing systems. Manufacturing Letters, 3, 18–23. https://doi.org/10.1016/j.mfglet.2014.12.001
  33. Lionberger, R. A., Lee, S. L., Lee, L. M., Raw, A., & Yu, L. X. (2008). Quality by design: Concepts for ANDAs. The AAPS Journal, 10(2), 268–276. https://doi.org/10.1208/s12248-008-9026-7
  34. Lundberg, S. M., & Lee, S.-I. (2017). A unified approach to interpreting model predictions. In Advances in neural information processing systems 30 (pp. 4765–4774).
  35. McMahan, H. B., Moore, E., Ramage, D., Hampson, S., & y Arcas, B. A. (2017). Communication-efficient learning of deep networks from decentralized data. In Proceedings of AISTATS 2017 (pp. 1273–1282).
  36. MHRA. (2021). GxP data integrity guidance and definitions (Revision). UK Medicines and Healthcare products Regulatory Agency.
  37. Nomikos, P., & MacGregor, J. F. (1995). Multivariate SPC charts for monitoring batch processes. Technometrics, 37(1), 41–59.
  38. https://doi.org/10.1080/00401706.1995.10484358
  39. PIC/S. (2021). PI 041-1: Good practices for data integrity in regulated GxP environments. Pharmaceutical Inspection Co-operation Scheme.
  40. PIC/S. (2023). PE 009-16: Guide to good manufacturing practice for medicinal products (with Annexes). Pharmaceutical Inspection Co-operation Scheme.
  41. Roggo, Y., Chalus, P., Maurer, L., Lema-Martinez, C., Edmond, A., & Jent, N. (2007). A review of near infrared spectroscopy and chemometrics in pharmaceutical technologies. Journal of Pharmaceutical and Biomedical Analysis, 44(3), 683–700. https://doi.org/10.1016/j.jpba.2007.03.023
  42. Rosen, R., Wichert, G., Lo, G., & Bettenhausen, K. D. (2015). About the importance of autonomy and digital twins for the future of manufacturing. In Proceedings of IEEE ETFA (pp. 1–8).
  43. Russell, S., & Norvig, P. (2021). Artificial intelligence: A modern approach (4th ed.). Pearson.
  44. Samek, W., Wiegand, T., & Müller, K.-R. (2017). Explainable artificial intelligence: Understanding, visualizing and interpreting deep learning models. IT Professional, 19(6), 38–46. https://doi.org/10.1109/MITP.2017.3122402
  45. Tao, F., Zhang, H., Liu, A., & Nee, A. Y. C. (2018). Digital twin in industry: State-of-the-art. IEEE Transactions on Industrial Informatics, 15(4), 2405–2415. https://doi.org/10.1109/TII.2018.2873186
  46. Touw, W. G., Bayjanov, J. R., Overmars, L., Backus, L., Boekhorst, J., & Wels, M. (2013). Data mining in the pharmaceutical industry: Enhancing PAT and QbD with chemometrics. Pharmaceutical Engineering, 33(6), 1–12.
  47. U.S. Code of Federal Regulations. (2024). 21 CFR Part 11—Electronic records; electronic signatures. U.S. Government Publishing Office.
  48. U.S. Code of Federal Regulations. (2024). 21 CFR Parts 210–211—Current good manufacturing practice for finished pharmaceuticals. U.S. Government Publishing Office.
  49. Vapnik, V. N. (1998). Statistical learning theory. Wiley.
  50. Vora, J., Bhadra, B., & Trivedi, K. (2020). Applications of machine learning in pharmaceutical product development and quality by design. International Journal of Pharmaceutical Sciences Review and Research, 65(1), 1–10.
  51. Wang, K., & Tsui, K.-L. (2018). Statistical process monitoring of complex manufacturing processes: With applications in pharmaceutical industry. Quality and Reliability Engineering International, 34(2), 175–189. https://doi.org/10.1002/qre.2224
  52. WHO. (2016). Good data and record management practices (Annex 5, WHO Technical Report Series, No. 996). World Health Organization.
  53. Woodcock, J. (2004). The concept of a quality systems approach to pharmaceutical CGMP regulations. Pharmaceutical Technology, 28(9), 68–71.
  54. Yu, L. X., Amidon, G., Khan, M. A., Hoag, S. W., Polli, J., Raju, G. K., & Woodcock, J. (2014). Understanding pharmaceutical quality by design. AAPS Journal, 16(4), 771–783. https://doi.org/10.1208/s12248-014-9598-3
  55. Zhang, L., Jiang, Z., & Wang, K. (2019). Deep learning-based visual inspection in manufacturing: A review. Journal of Manufacturing Systems, 54, 258–276. https://doi.org/10.1016/j.jmsy.2019.01.007
  56. Zou, H., & Hastie, T. (2005). Regularization and variable selection via the elastic net. Journal of the Royal Statistical Society: Series B, 67(2), 301–320. https://doi.org/10.1111/j.1467-9868.2005.00503.x

Reference

  1. STM International. (2020). ASTM E2500-20: Standard guide for specification, design, and verification of pharmaceutical and biopharmaceutical manufacturing systems and equipment. ASTM International. https://doi.org/10.1520/E2500-20
  2. ASTM International. (2017). ASTM E2968-17: Standard guide for application of continuous process verification to pharmaceutical and biopharmaceutical manufacturing. ASTM International.
  3. Bakeev, K. A. (Ed.). (2010). Process analytical technology: Spectroscopic tools and implementation strategies for the chemical and pharmaceutical industries (2nd ed.). Wiley. https://doi.org/10.1002/9780470689592
  4. Bishop, C. M. (2006). Pattern recognition and machine learning. Springer.
  5. Breiman, L. (2001). Random forests. Machine Learning, 45(1), 5–32. https://doi.org/10.1023/A:1010933404324
  6. Carvalho, T. P., Soares, F. A. A. M. N., Vita, R., Francisco, R. de P., Basto, J. P., & Alcalá, S. G. S. (2019). A systematic literature review of machine learning methods applied to predictive maintenance. Computers & Industrial Engineering, 137, 106024. https://doi.org/10.1016/j.cie.2019.106024
  7. Casino, F., Dasaklis, T. K., & Patsakis, C. (2019). A systematic literature review of blockchain-based applications: Current status, classification and open issues. Telematics and Informatics, 36, 55–81. https://doi.org/10.1016/j.tele.2018.11.006
  8. Cortes, C., & Vapnik, V. (1995). Support-vector networks. Machine Learning, 20(3), 273–297. https://doi.org/10.1007/BF00994018
  9. Devlin, J., Chang, M.-W., Lee, K., & Toutanova, K. (2019). BERT: Pre-training of deep bidirectional transformers for language understanding. In Proceedings of NAACL-HLT 2019 (pp. 4171–4186).
  10. Doshi-Velez, F., & Kim, B. (2017). Towards a rigorous science of interpretable machine learning. arXiv. https://arxiv.org/abs/1702.08608
  11. European Commission. (2022). EudraLex Volume 4: EU Guidelines for Good Manufacturing Practice for Medicinal Products for Human and Veterinary Use—Annex 1: Manufacture of sterile medicinal products (Revision).
  12. European Medicines Agency. (2011). Annex 11: Computerised systems (EU GMP, Volume 4).
  13. European Medicines Agency. (2012). Guideline on real time release testing (RTRT) of finished products. EMA/CHMP/QWP/811210/2009.
  14. Food and Drug Administration. (2004). Guidance for industry: PAT—A framework for innovative pharmaceutical development, manufacturing, and quality assurance. U.S. Department of Health and Human Services.
  15. Food and Drug Administration. (2011). Guidance for industry: Process validation—General principles and practices. U.S. Department of Health and Human Services.
  16. Food and Drug Administration. (2018). Data integrity and compliance with drug CGMP: Questions and answers—Guidance for industry. U.S. Department of Health and Human Services.
  17. Food and Drug Administration (CDER & CBER). (2023). Discussion paper: Using artificial intelligence & machine learning in the development of drug & biological products. U.S. Food and Drug Administration.
  18. Goodfellow, I., Bengio, Y., & Courville, A. (2016). Deep learning. MIT Press.
  19. Guidotti, R., Monreale, A., Ruggieri, S., Turini, F., Giannotti, F., & Pedreschi, D. (2018). A survey of methods for explaining black box models. ACM Computing Surveys, 51(5), 93. https://doi.org/10.1145/3236009
  20. ICH. (2008). ICH Q10: Pharmaceutical quality system. International Council for Harmonisation.
  21. ICH. (2009). ICH Q8(R2): Pharmaceutical development. International Council for Harmonisation.
  22. ICH. (2023). ICH Q9(R1): Quality risk management (Revision 1). International Council for Harmonisation.
  23. ICH. (2022). ICH Q13: Continuous manufacturing of drug substances and drug products. International Council for Harmonisation.
  24. ICH. (2023). ICH Q14: Analytical procedure development. International Council for Harmonisation.
  25. ISPE. (2017). GAMP® Guide: Records and data integrity. International Society for Pharmaceutical Engineering.
  26. ISPE. (2022). GAMP® 5, Second Edition: A risk-based approach to compliant GxP computerized systems. International Society for Pharmaceutical Engineering.
  27. Jardine, A. K. S., Lin, D., & Banjevic, D. (2006). A review on machinery diagnostics and prognostics implementing condition-based maintenance. Mechanical Systems and Signal Processing, 20(7), 1483–1510. https://doi.org/10.1016/j.ymssp.2005.09.012
  28. Jurafsky, D., & Martin, J. H. (2023). Speech and language processing (3rd ed., draft). Stanford University/Prentice Hall.
  29. Kone?ný, J., McMahan, H. B., Yu, F. X., Richtárik, P., Suresh, A. T., & Bacon, D. (2016). Federated learning: Strategies for improving communication efficiency. arXiv. https://arxiv.org/abs/1610.05492
  30. Kourti, E. (2005). Application of latent variable methods to process control and multivariate statistical process control in industry. International Journal of Adaptive Control and Signal Processing, 19(4), 213–246. https://doi.org/10.1002/acs.859
  31. Kuhn, M., & Johnson, K. (2013). Applied predictive modeling. Springer. https://doi.org/10.1007/978-1-4614-6849-3
  32. Lee, J., Bagheri, B., & Kao, H.-A. (2015). A cyber-physical systems architecture for industry 4.0-based manufacturing systems. Manufacturing Letters, 3, 18–23. https://doi.org/10.1016/j.mfglet.2014.12.001
  33. Lionberger, R. A., Lee, S. L., Lee, L. M., Raw, A., & Yu, L. X. (2008). Quality by design: Concepts for ANDAs. The AAPS Journal, 10(2), 268–276. https://doi.org/10.1208/s12248-008-9026-7
  34. Lundberg, S. M., & Lee, S.-I. (2017). A unified approach to interpreting model predictions. In Advances in neural information processing systems 30 (pp. 4765–4774).
  35. McMahan, H. B., Moore, E., Ramage, D., Hampson, S., & y Arcas, B. A. (2017). Communication-efficient learning of deep networks from decentralized data. In Proceedings of AISTATS 2017 (pp. 1273–1282).
  36. MHRA. (2021). GxP data integrity guidance and definitions (Revision). UK Medicines and Healthcare products Regulatory Agency.
  37. Nomikos, P., & MacGregor, J. F. (1995). Multivariate SPC charts for monitoring batch processes. Technometrics, 37(1), 41–59.
  38. https://doi.org/10.1080/00401706.1995.10484358
  39. PIC/S. (2021). PI 041-1: Good practices for data integrity in regulated GxP environments. Pharmaceutical Inspection Co-operation Scheme.
  40. PIC/S. (2023). PE 009-16: Guide to good manufacturing practice for medicinal products (with Annexes). Pharmaceutical Inspection Co-operation Scheme.
  41. Roggo, Y., Chalus, P., Maurer, L., Lema-Martinez, C., Edmond, A., & Jent, N. (2007). A review of near infrared spectroscopy and chemometrics in pharmaceutical technologies. Journal of Pharmaceutical and Biomedical Analysis, 44(3), 683–700. https://doi.org/10.1016/j.jpba.2007.03.023
  42. Rosen, R., Wichert, G., Lo, G., & Bettenhausen, K. D. (2015). About the importance of autonomy and digital twins for the future of manufacturing. In Proceedings of IEEE ETFA (pp. 1–8).
  43. Russell, S., & Norvig, P. (2021). Artificial intelligence: A modern approach (4th ed.). Pearson.
  44. Samek, W., Wiegand, T., & Müller, K.-R. (2017). Explainable artificial intelligence: Understanding, visualizing and interpreting deep learning models. IT Professional, 19(6), 38–46. https://doi.org/10.1109/MITP.2017.3122402
  45. Tao, F., Zhang, H., Liu, A., & Nee, A. Y. C. (2018). Digital twin in industry: State-of-the-art. IEEE Transactions on Industrial Informatics, 15(4), 2405–2415. https://doi.org/10.1109/TII.2018.2873186
  46. Touw, W. G., Bayjanov, J. R., Overmars, L., Backus, L., Boekhorst, J., & Wels, M. (2013). Data mining in the pharmaceutical industry: Enhancing PAT and QbD with chemometrics. Pharmaceutical Engineering, 33(6), 1–12.
  47. U.S. Code of Federal Regulations. (2024). 21 CFR Part 11—Electronic records; electronic signatures. U.S. Government Publishing Office.
  48. U.S. Code of Federal Regulations. (2024). 21 CFR Parts 210–211—Current good manufacturing practice for finished pharmaceuticals. U.S. Government Publishing Office.
  49. Vapnik, V. N. (1998). Statistical learning theory. Wiley.
  50. Vora, J., Bhadra, B., & Trivedi, K. (2020). Applications of machine learning in pharmaceutical product development and quality by design. International Journal of Pharmaceutical Sciences Review and Research, 65(1), 1–10.
  51. Wang, K., & Tsui, K.-L. (2018). Statistical process monitoring of complex manufacturing processes: With applications in pharmaceutical industry. Quality and Reliability Engineering International, 34(2), 175–189. https://doi.org/10.1002/qre.2224
  52. WHO. (2016). Good data and record management practices (Annex 5, WHO Technical Report Series, No. 996). World Health Organization.
  53. Woodcock, J. (2004). The concept of a quality systems approach to pharmaceutical CGMP regulations. Pharmaceutical Technology, 28(9), 68–71.
  54. Yu, L. X., Amidon, G., Khan, M. A., Hoag, S. W., Polli, J., Raju, G. K., & Woodcock, J. (2014). Understanding pharmaceutical quality by design. AAPS Journal, 16(4), 771–783. https://doi.org/10.1208/s12248-014-9598-3
  55. Zhang, L., Jiang, Z., & Wang, K. (2019). Deep learning-based visual inspection in manufacturing: A review. Journal of Manufacturing Systems, 54, 258–276. https://doi.org/10.1016/j.jmsy.2019.01.007
  56. Zou, H., & Hastie, T. (2005). Regularization and variable selection via the elastic net. Journal of the Royal Statistical Society: Series B, 67(2), 301–320. https://doi.org/10.1111/j.1467-9868.2005.00503.x

Photo
Divya Deshmukh
Corresponding author

Samarth Institute of Pharmacy Belhe, Pune, Maharashtra, India.

Photo
Dr. Sachin Datkhile
Co-author

Samarth Institute of Pharmacy Belhe, Pune, Maharashtra, India.

Photo
Omkar Gosavi
Co-author

Samarth Institute of Pharmacy Belhe, Pune, Maharashtra, India.

Photo
Komal Aher
Co-author

Samarth Institute of Pharmacy Belhe, Pune, Maharashtra, India.

Photo
Komal Gaikwad
Co-author

Samarth Institute of Pharmacy Belhe, Pune, Maharashtra, India.

Photo
Bhakti Gurav
Co-author

Samarth Institute of Pharmacy Belhe, Pune, Maharashtra, India.

Divya Deshmukh, Dr. Sachin Datkhile, Omkar Gosavi, Komal Aher, Komal Gaikwad, Bhakti Gurav, Artificial Intelligence and Machine Learning in Pharmaceutical Quality Assurance, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 9, 771-786. https://doi.org/10.5281/zenodo.17067967

More related articles
Formulation And Evaluation Of Tea Powder From Fenu...
Avantika .R. Dongare , Pankaj Vyawhare, ...
UV Spectroscopic Estimation of Brivaracetam in Bul...
Dr Kumar Raja Jayavarapu, M. Manikanta, M. Dharani, M. Mani Likit...
Related Articles
Quality Assurance in Pharmaceutical Manufacturing : Ensuring Consistency and Saf...
Sachin Chalge, Vinod Chaware, Tejas Sharma, Dr. Shivshankar D. Mhaske, Sanket Kakade, ...
Exploring The Anti-Anaemic Potential of Medicinal Plants Found in Uttarakhand: A...
Chandrakanta, Shashi Bisht, Nikita Bankoti, Pushpendra Yadav , ...
Herbal Remedy for Stomach Pain: Formulation and Efficacy...
Kadam Bhagyashri , Shete Pooja , Aher Aniruddha , Sabale J. V., ...
Formulation And Evaluation Of Herbal Vitamin C Tablet...
Abhishek A. Adhav, Abhishek Uttam Gaikwad, Abhijeet R. Shete, Megha T. Salve , ...