View Article

  • Improvement in Designing, Formulation, and Development through QBD-Based SOPs: A Review

  • Roorkee College of Pharmacy, Dist. Haridwar, Uttarakhand.

Abstract

The landscape of pharmaceutical development has transitioned from conventional trial-and-error methods to more structured, science-driven, and risk-based approaches. Among these, Quality by Design (QbD) has established itself as a pivotal framework in optimizing formulation and manufacturing processes. Integrating QbD principles into Standard Operating Procedures (SOPs) fosters a systematic and data-centric pathway for product development, ensuring enhanced reproducibility and regulatory compliance. By embedding quality at the earliest stages, QbD-based SOPs contribute to improved formulation strategies, reduced variability, and better alignment with global regulatory expectations. This review highlights the critical role of QbD-integrated SOPs in strengthening pharmaceutical design, advancing formulation robustness, and streamlining development activities. Key elements such as Quality Target Product Profile (QTPP), Critical Quality Attributes (CQAs), Critical Process Parameters (CPPs), and the application of risk management methodologies are explored. In addition, practical insights through case studies and specific applications within the Indian pharmaceutical sector are discussed to illustrate the real-world impact of QbD-based SOPs. It highlights key QbD elements—Quality Target Product Profile (QTPP), Critical Quality Attributes (CQAs), Critical Material Attributes (CMAs), and Critical Process Parameters (CPPs)—and explores their incorporation into SOPs to achieve regulatory compliance and product quality assurance.

Keywords

Quality by Design (QBD), Standard Operating Procedures (SOPs), pharmaceutical formulation, risk assessment, process development, regulatory compliance

Introduction

In the evolving landscape of pharmaceutical development, ensuring product quality, safety, and therapeutic efficacy remains a fundamental priority. Traditional methods, heavily dependent on empirical trial-and-error approaches, often led to prolonged development timelines and inconsistencies in product quality. The introduction of Quality by Design (QbD), as outlined in the ICH Q8 (R2) guideline, has marked a significant departure from the conventional quality-by-testing model towards a more proactive quality-by-design philosophy. By embedding QbD principles within Standard Operating Procedures (SOPs), a structured, knowledge-driven framework for drug development can be established.1 The pharmaceutical industry is increasingly transitioning from empirical strategies to systematic, science- and risk-based development models. Quality by Design (QbD) has emerged as a central strategy to ensure quality, consistency, and regulatory conformity throughout the drug development lifecycle. Regulatory bodies such as the FDA and the International Council for Harmonisation (ICH) advocate for QbD implementation, which is rooted in a deep scientific understanding of critical quality attributes (CQAs), critical material attributes (CMAs), and critical process parameters (CPPs). This proactive approach enables more effective risk management and enhances overall product quality.2,3 Within this context, Standard Operating Procedures (SOPs) become instrumental in operationalizing QbD concepts into day-to-day pharmaceutical development practices. By integrating scientific principles and risk assessments into SOPs, pharmaceutical companies can develop more robust and reproducible processes across formulation, manufacturing, and quality control activities.4 This integration minimizes process variability, enhances efficiency, and reduces the likelihood of product failure. The incorporation of QbD into SOPs not only optimizes formulation and development strategies but also supports data-driven decision-making through the use of tools like Design of Experiments (DoE) and risk assessment methodologies.5 Moreover, it fosters a culture of continuous improvement and regulatory flexibility, essential for staying competitive in the global pharmaceutical market.6 This review delves into the pivotal role of QbD in advancing pharmaceutical product design, formulation, and development through the lens of SOP-driven practices. It aims to illustrate how combining structured documentation with a profound scientific understanding of processes leads to improved product quality, shortened development timelines, and enhanced compliance with international regulatory standards.7

2. The Concept and Evolution of QbD

Quality by Design (QbD) represents a systematic, proactive framework for pharmaceutical development, beginning with clearly defined objectives. It emphasizes deep process understanding, scientific reasoning, and robust risk management to ensure consistent product quality. Unlike traditional quality control approaches that rely heavily on end-product testing, QbD seeks to embed quality into the product from the earliest stages of development.8,9

2.1. Origins and Regulatory Foundation

The foundational ideas behind QbD were first introduced in the manufacturing industry by Dr. Joseph M. Juran as part of his broader philosophy of quality management.10,11 In the pharmaceutical sector, QbD began gaining significant traction following the release of the U.S. FDA's Pharmaceutical CGMPs for the 21st Century – A Risk-Based Approach (2004). Its principles were subsequently reinforced through a series of International Council for Harmonisation (ICH) guidelines, notably:

  • ICH Q8 (R2): Pharmaceutical Development
  • ICH Q9: Quality Risk Management
  • ICH Q10: Pharmaceutical Quality System.12

These regulatory frameworks collectively advocated a shift away from conventional quality-by-testing models toward a more deliberate, design-driven quality paradigm.12

2.2. Core Principles of QbD

Implementing QbD involves several essential components:

  • Quality Target Product Profile (QTPP): A prospective summary outlining the critical quality characteristics the final product must possess.
  • Critical Quality Attributes (CQAs): Specific physical, chemical, biological, or microbiological properties that must be maintained within acceptable limits to ensure product quality.
  • Critical Material Attributes (CMAs) and Critical Process Parameters (CPPs): Key material properties and process parameters that directly impact the CQAs and overall product performance.
  • Design Space: The established range of input variables and process parameters that assure consistent quality.
  • Control Strategy: A comprehensive plan comprising controls based on product and process understanding.
  • Lifecycle Management: A continuous improvement approach where knowledge gained throughout the product's lifecycle informs future development and control strategies.13

2.3. Evolution and Industrial Adoption

While the initial reception of QbD was cautious—largely due to the paradigm shift it demanded from traditional practices—it has steadily gained global acceptance over the past decade.14 With enhanced regulatory support and clearer technical guidance, pharmaceutical companies have increasingly integrated QbD into both new drug development and post-approval product changes. Advanced tools such as Design of Experiments (DoE),15 risk assessment methodologies, Process Analytical Technology (PAT),16 and integrated knowledge management17 systems have played critical roles in strengthening the pharmaceutical development pipeline.

Today, QbD is recognized not merely as a regulatory requirement but as a strategic advantage. It enables more robust process design, deeper product understanding, and ultimately contributes to the delivery of safer, more effective therapies to patients worldwide.18

3. Essential Part of QbD

Element

Description

QTPP

Identifies target characteristics of a product

CQA

Attributes that must be controlled to ensure product quality

CMA

Material properties affecting CQAs

CPP

Process variables influencing CQAs

Control Strategy

Set of controls to ensure product quality

Design Space

Multidimensional space that ensures product quality

4. Standard Operating Procedures (SOPs) in Pharmaceutical Development

Standard Operating Procedures (SOPs) are comprehensive, documented instructions intended to standardize the execution of specific tasks. In the context of pharmaceutical development, SOPs form the foundation of operational compliance, governing activities across raw material management, formulation processes, analytical testing, packaging, and regulatory documentation.19

4.1. Importance of SOPs in a Regulated Environment

Operating within a highly regulated framework, the pharmaceutical industry must comply with standards set by authorities such as the U.S. Food and Drug Administration (FDA), European Medicines Agency (EMA), and Central Drugs Standard Control Organization (CDSCO).SOPs play a critical role in ensuring that operations consistently adhere to Good Manufacturing Practices (GMP), Good Laboratory Practices (GLP), and Good Clinical Practices (GCP).20 Beyond maintaining compliance, SOPs minimize the risk of human error, uphold accountability, and ensure traceability throughout the development and manufacturing continuum.21

4.2. SOPs as Vehicles for QbD Implementation

Embedding Quality by Design (QbD) principles into SOPs elevates them from static procedural documents to dynamic instruments of quality assurance. QbD-integrated SOPs facilitate:

  • Systematic definition and documentation of Quality Target Product Profiles (QTPPs), Critical Quality Attributes (CQAs), Critical Process Parameters (CPPs), and Critical Material Attributes (CMAs).
  • Structured application of Design of Experiments (DoE) for formulation development and process optimization.
  • Consistent execution of risk assessments and documentation of control strategies. Enhanced knowledge transfer across research, development, and manufacturing teams throughout the product lifecycle. By incorporating QbD frameworks, SOPs support science-driven, risk-based decision-making, ultimately strengthening the reliability and predictability of pharmaceutical processes.22,23

4.3. SOPs across the Product Development Lifecycle

SOPs are integral at each stage of drug development:

  • Pre-formulation and Formulation Design: Guiding physicochemical assessments, excipient selection, and initial DoE planning.24
  • Process Development and Scale-Up: Directing the evaluation of process parameters, pilot batch production, and transition to larger-scale manufacturing.
  • Analytical Method Development: Ensuring the creation and validation of accurate, reproducible analytical methods for both active pharmaceutical ingredients (APIs) and finished products.
  • Technology Transfer and Commercial Manufacturing: Facilitating the smooth transfer of processes from research and development (R&D) to production units while maintaining quality standards and regulatory compliance.25

4.4. Digitalization and Continuous Improvement

The management of SOPs is increasingly transitioning to Electronic Document Management Systems (EDMS), which offer advantages such as version control, audit trail maintenance, and seamless integration with broader Quality Management Systems (QMS). When paired with real-time process monitoring tools and Process Analytical Technology (PAT),  digital SOPs support Continuous Process Verification (CPV) and lifecycle management, key aspects of a robust QbD strategy.

5. Integrating QbD with SOPs: Strategic Approaches

Achieving successful integration of Quality by Design (QbD) into Standard Operating Procedures (SOPs) demands a structured and strategic methodology. Such alignment not only ensures regulatory compliance but also enhances product quality, operational consistency, and lifecycle management.

5.1. Structuring SOPs Based on QbD Components

For SOPs to effectively reflect QbD principles, their structure must correspond with key QbD elements:

  • Quality Target Product Profile (QTPP): SOPs should include designated sections or templates for defining the QTPP based on therapeutic goals and patient-centric needs.6
  • Critical Quality Attributes (CQAs): Specific procedures should guide the systematic identification, evaluation, and justification of CQAs for various dosage forms.6
  • Critical Material Attributes (CMAs) and Critical Process Parameters (CPPs): SOPs must mandate risk assessments and the implementation of control strategies addressing material variability and process fluctuations.
  • Design Space Development: Procedures should outline steps for establishing, documenting, and justifying the design space using scientific data and Design of Experiments (DoE) methodologies.26

5.2. Cross-Functional Collaboration in SOP Development

Integrating QbD into SOPs requires a multidisciplinary approach, involving Research and Development (R&D), Quality Assurance (QA), Regulatory Affairs, and Manufacturing departments. Active collaboration ensures that SOPs encapsulate comprehensive process insights and remain applicable across different stages of product development and commercialization.

5.3. Incorporation of Risk Assessment Tools

To support a QbD-centric framework, SOPs should embed risk management tools, including:

  • Failure Mode and Effects Analysis (FMEA)
  • Ishikawa (Fishbone) Diagrams
  • Risk Priority Number (RPN) Scoring

Embedding these tools into routine SOP processes aids in systematically identifying potential failure modes, prioritizing risks, and establishing effective control measures.

5.4. Dynamic Documentation and Continuous Feedback

SOPs should be treated as dynamic, evolving documents. Regular updates based on process performance metrics, regulatory changes, and post-approval modifications are vital. Integrating feedback mechanisms—such as Corrective and Preventive Action (CAPA) systems—into SOP workflows enables continuous refinement and promotes proactive quality improvement.

5.5. Training and Implementation Strategies

Training is a critical success factor for the effective implementation of QbD-based SOPs. Personnel must be educated not only on procedural steps but also on the underlying scientific rationale behind QbD elements. Such training enhances process understanding, fosters a quality-centric culture, and ensures uniformity in SOP execution across functional teams.27

Flowchart: Integration of QbD into SOP Development Lifecycle

6. Practical Applications and Case Studies in India

The practical application of QbD-based Standard Operating Procedures (SOPs) in pharmaceutical development has resulted in notable advancements in product quality, process robustness, and regulatory compliance. Various case studies from the Indian pharmaceutical landscape illustrate how systematic integration of QbD elements into SOPs can enhance drug development and manufacturing outcomes.

6.1. Case Study: Development of an Extended-Release Metoprolol Succinate Tablet

Organization: Multinational Pharmaceutical Company
Product: Extended-release tablet of Metoprolol Succinate
Objective: Develop a robust formulation achieving both in vitro and in vivo bioequivalence standards.

QBD-SOP Integration:

  • The Quality Target Product Profile (QTPP) was outlined based on desired release kinetics and target plasma concentration profiles.
  • Design of Experiments (DoE) was employed to assess the influence of polymer concentration, granule size, and compression force on Critical Quality Attributes (CQAs).5,30
  • SOPs governing granulation and compression operations were updated to incorporate optimized material attributes and critical process parameters.
  • A Design Space was established and formally documented within SOPs.

The optimized formulation demonstrated consistent performance across multiple batches, significantly reduced the need for rework, and achieved expedited regulatory approval.25, 26, 28

6.2. Case Study: Risk-Based Process Validation for a Parenteral Ceftriaxone Product

Organization: Medium-Sized Injectable Manufacturing Company
Product: Injectable Ceftriaxone Sodium
Challenge: High inter-batch variability and Out-of-Specification (OOS) occurrences due to sterility assurance failures.

QBD-SOP Strategy:

  • SOPs were updated to include Failure Mode and Effects Analysis (FMEA)-based risk assessments for critical unit operations (e.g., filtration, aseptic filling, terminal sterilization).13,24
  • Identification of Critical Process Parameters (CPPs), including sterilization temperatures, fill volume accuracy, and filter integrity performance.
  • Deployment of Process Analytical Technology (PAT) tools for real-time critical parameter monitoring.17
  • Strengthened documentation practices and preventive maintenance SOPs to mitigate human error.19

6.3. Application Example: QbD-Enabled Analytical Method Development for a Poorly Soluble Drug

Development of an HPLC method for a poorly water-soluble Active Pharmaceutical Ingredient (API).

SOP Enhancement:

  • Inclusion of a structured DoE-based approach within SOPs for analytical method development, focusing on factors such as mobile phase composition, pH, and detection wavelength.
  • Establishment of system suitability criteria (e.g., resolution, tailing factor) as part of the defined CQAs.
  • Upgradation of method validation SOPs to incorporate robustness testing and lifecycle management principles.29,30

6.4. Pilot Study: Implementing QbD-Driven SOPs Across a Solid Dosage Manufacturing Plant:-

Scope: Integration of QbD concepts into SOPs across five functional departments—Research & Development (R&D), Quality Assurance (QA), Quality Control (QC), Production, and Regulatory Affairs.

Implement measures

  • Development of standardized QTPP templates and risk assessment forms.
  • Revision and alignment of over 35 SOPs to incorporate QbD frameworks.
  • Comprehensive training programs delivered to staff focusing on updated documentation practices and the scientific basis of QbD.6,19

Benefits:

  • Achieved a 30% reduction in batch failure rates.
  • Improved readiness for regulatory audits and accelerated regulatory submission timelines.
  • Strengthened interdepartmental communication and promoted a culture of continuous knowledge sharing.

Case Study

Organization Type

Product

QbD-SOP Approach

Key Outcomes

Development of Extended-Release Tablet

Multinational Pharmaceutical Company

Metoprolol Succinate Extended-Release Tablets

- Defined QTPP and CQAs
- Used DoE to optimize formulation parameters
- Updated SOPs to document design space and process controls

- Consistent batch performance
- Reduced rework
- Faster regulatory approval

Risk-Based Process Validation for Injectable

Medium-Sized Injectable Manufacturer

Parenteral Ceftriaxone Sodium

- FMEA-based risk assessments in SOPs
- CPPs identification and PAT integration
- Improved preventive maintenance SOPs

- >80% reduction in sterility-related deviations
- Improved batch release timelines

QbD-Driven Analytical Method Development

Pharmaceutical R&D Scenario

Poorly Soluble API (HPLC method development)

- SOPs included DoE-driven method development
- System suitability parameters as CQAs
- Validation SOP included robustness criteria

- High method precision and robustness
- Successful method transfer across labs

Pilot Study for QbD-SOP Implementation in Plant

Solid Dosage Form Plant

Multiple Solid Dosage Products

- Developed QTPP templates and risk assessment forms
- Revised 35+ SOPs
- Staff training on scientific rationale of QbD

- 30% reduction in batch failures
- Enhanced audit readiness
- Faster regulatory submission

Benefits from Case Studies

Parameter

Before QbD-SOP Integration

After QbD-SOP Integration

Batch Variability

High

Significantly reduced

Regulatory Query Response Time

Slow and inconsistent

Faster, with supporting data

Product Understanding

Empirical and fragmented

Holistic and science-based

Change Management

Reactive

Proactive and risk-informed

Compliance During Audits

Challenging

Improved and well-documented

7. Benefits of QbD-Based SOPs

Benefit

Impact

Robust Design

Greater product consistency and control

Reduced Development Time

Efficient experimental planning

Regulatory Acceptance

Facilitates ANDA/NDA approval

Cost Savings

Less rework and batch failure

Knowledge Sharing

Encourages documentation and learning

8. Challenges in Implementation

While QbD-based SOPs significantly enhance pharmaceutical development, their implementation presents several challenges:

Challenge

Description

Impact

Mitigation Strategy

High Initial Resource Requirement

Significant investment in time, expertise, and advanced tools is needed.

Barrier for small or mid-sized firms.

Phased implementation focusing on high-impact areas.

Complexity in SOP Design and Maintenance

SOPs become more detailed and technically dense.

Risk of errors, information overload, difficulty in version control.

Modular SOP design and use of document management systems.

Resistance to Change

Employees may be hesitant to adopt new practices.

Slow adoption, inconsistency in application.

Regular training and change management initiatives.

Data Management and Documentation Burden

Large volumes of data need organized storage and analysis.

Increased administrative workload, potential data loss.

Use of EDMS and knowledge management platforms.

Regulatory Ambiguity

Variability in QbD expectations across global regulatory agencies.

Uncertainty in submission requirements.

Early regulatory dialogue and clarification meetings.

Limited Integration with Legacy Systems

Difficulty merging QbD into existing workflows.

Risk of process disruption or duplication of efforts.

Strategic planning and gradual system upgrades.

Over-Reliance on Tools

Risk of mechanical application without true scientific understanding.

Loss of the true spirit of QbD.

Emphasis on scientific training and rational thinking beyond tools.

9. CONCLUSION

The integration of Quality by Design (QbD) principles into Standard Operating Procedures (SOPs) represents a transformative shift in pharmaceutical development and manufacturing. By embedding scientific understanding, risk-based thinking, and process control strategies into standardized workflows, QbD-based SOPs enhance not only product quality but also efficiency, consistency, and regulatory compliance. This review highlights how QbD-aligned SOPs serve as structured blueprints that guide formulation scientists, process engineers, and quality professionals in making informed, data-driven decisions. From defining the Quality Target Product Profile (QTPP) to establishing control strategies and managing lifecycle changes, these SOPs act as operational manifestations of pharmaceutical quality systems. Despite challenges such as resource intensiveness, data complexity, and the need for cultural adaptation, the long-term benefits of QbD-based SOPs are significant. These include improved batch-to-batch consistency, faster product development, fewer deviations, and enhanced readiness for regulatory inspections.

REFERENCES

  1. ICH Q8 (R2) – Pharmaceutical Development. International Conference on Harmonisation.
  2. CDSCO (India). Guidance for Industry on Pharmaceutical Development Using QbD Approach.
  3. Jha, A. K., & Singh, R. K. (2020). Implementation of QbD Approach. Journal of Drug Delivery and Therapeutics, 10(6), 153-158.
  4. Patel, B. A., & Shah, D. A. (2019). QbD: A Scientific Tool. Int. J. Pharm. Sci. Rev. Res., 59(1), 1–6.
  5. Sahu, S., & Panda, S. K. (2021). Role of QbD in SOPs. Indian J. Pharm. Sci. Res., 11(3), 102–108.
  6. Dureja, H., & Saini, V. (2017). QbD in Pharmaceutical Development. Pharma Times, 49(5), 24–27.
  7.  Aulton, M. E., & Taylor, K. M. G. (2017). Aulton’s Pharmaceutics, 5th Ed., Elsevier.
  8. Subrahmanyam, C. V. S. (2020). Pharmaceutical Production and Management, 2nd Ed., Vallabh Prakashan.
  9.  FDA (USA). Guidance for Industry: QbD in ANDA Submissions.
  10. Kaur, R., et al. (2021). Application of QbD in Formulation Development. Asian J. Pharm. Clin. Res., 14(1), 45–50.
  11. Sharma, A., & Gupta, R. (2020). Application of QbD in Development of Novel Drug Delivery Systems. Int. J. Pharm. Sci. Drug Res., 12(2), 85–90.
  12. Rao, M., & Kulkarni, R. (2018). Design of Experiments for Pharmaceutical Process Optimization. Indian J. Pharm. Educ. Res., 52(3), 310–316.
  13. Bhalodia, R., & Patel, G. (2017). Quality by Design Approach in Formulation Development. Pharmatutor, 5(12), 20–25.
  14. Kumar, A., et al. (2021). Role of QbD in Pharmaceutical Industry. J. Pharm. Sci. & Res., 13(5), 270–274.
  15. Bansal, A. K. (2016). Pharmaceutical Quality by Design: A Practical Approach. Pharmabiz.com.
  16. Chaturvedi, R., & Dubey, R. (2019). Implementation of QbD Principles in Pharmaceutical Manufacturing. Int. J. Drug Dev. & Res., 11(1), 100–105.
  17. Singh, P., & Verma, R. (2020). Risk Assessment Tools in QbD: A Comprehensive Review. Int. J. Pharm. Sci. Invent., 9(6), 45–50.
  18. Joshi, R. D., & Mehta, T. A. (2018). A Review on Quality by Design (QbD). Int. J. Adv. Pharm., 7(1), 32–37.
  19. Desai, R. J., & Shah, D. R. (2022). Benefits and Challenges of QbD in the Indian Pharmaceutical Sector. Indian Drugs, 59(4), 12–18.
  20. Bhatt, D. A., & Rane, S. I. (2011). QbD: A Systematic Approach to Pharmaceutical Development. Pharma Times, 43(6), 16–20.
  21. Hinrichs WLJ, Möschwitzer JP. Quality by Design Approaches for Oral Drug Delivery Formulations. Adv Drug Deliv Rev. 2016;95:27–39.
  22. Raju NJ, et al. Challenges in Implementing QbD in Pharmaceutical Industries. Int J Pharm Sci Rev Res. 2014;28(1):72–78.
  23. Bolton S, Bon C. Pharmaceutical Statistics: Practical and Clinical Applications. 5th ed. CRC Press; 2009.
  24. Bhutani H, et al. Risk Assessment Tools in Pharmaceutical QbD. Pharm Anal Acta. 2014;5(6):1–8.
  25. Juran JM. Juran on Quality by Design: The New Steps for Planning Quality into Goods and Services. The Free Press; 1992.
  26. Narang AS, Desai D, Badawy S. Impact of Excipient Selection on Quality by Design (QbD) in Drug Product Development. AAPS PharmSciTech. 2012;13(3):1208–1217.
  27. Chow SC, Liu JP. Design and Analysis of Clinical Trials: Concepts and Methodologies. 3rd ed. Wiley; 2013.
  28. Xie W, Ji S, et al. Development of Controlled Release Systems Using QbD Approach. Eur J Pharm Biopharm. 2015;92:1–13.
  29. Kaur A, Kaur G. Application of QbD in Analytical Method Development: A Review. Int J Pharm Chem Biol Sci. 2013;3(4):1235–1244.
  30. Ferreiro I, et al. The Use of DoE in Pharmaceutical Development: An Industrial Perspective. Drug Dev Ind Pharm. 2020;46(7):1091–1103.

Reference

  1. ICH Q8 (R2) – Pharmaceutical Development. International Conference on Harmonisation.
  2. CDSCO (India). Guidance for Industry on Pharmaceutical Development Using QbD Approach.
  3. Jha, A. K., & Singh, R. K. (2020). Implementation of QbD Approach. Journal of Drug Delivery and Therapeutics, 10(6), 153-158.
  4. Patel, B. A., & Shah, D. A. (2019). QbD: A Scientific Tool. Int. J. Pharm. Sci. Rev. Res., 59(1), 1–6.
  5. Sahu, S., & Panda, S. K. (2021). Role of QbD in SOPs. Indian J. Pharm. Sci. Res., 11(3), 102–108.
  6. Dureja, H., & Saini, V. (2017). QbD in Pharmaceutical Development. Pharma Times, 49(5), 24–27.
  7.  Aulton, M. E., & Taylor, K. M. G. (2017). Aulton’s Pharmaceutics, 5th Ed., Elsevier.
  8. Subrahmanyam, C. V. S. (2020). Pharmaceutical Production and Management, 2nd Ed., Vallabh Prakashan.
  9.  FDA (USA). Guidance for Industry: QbD in ANDA Submissions.
  10. Kaur, R., et al. (2021). Application of QbD in Formulation Development. Asian J. Pharm. Clin. Res., 14(1), 45–50.
  11. Sharma, A., & Gupta, R. (2020). Application of QbD in Development of Novel Drug Delivery Systems. Int. J. Pharm. Sci. Drug Res., 12(2), 85–90.
  12. Rao, M., & Kulkarni, R. (2018). Design of Experiments for Pharmaceutical Process Optimization. Indian J. Pharm. Educ. Res., 52(3), 310–316.
  13. Bhalodia, R., & Patel, G. (2017). Quality by Design Approach in Formulation Development. Pharmatutor, 5(12), 20–25.
  14. Kumar, A., et al. (2021). Role of QbD in Pharmaceutical Industry. J. Pharm. Sci. & Res., 13(5), 270–274.
  15. Bansal, A. K. (2016). Pharmaceutical Quality by Design: A Practical Approach. Pharmabiz.com.
  16. Chaturvedi, R., & Dubey, R. (2019). Implementation of QbD Principles in Pharmaceutical Manufacturing. Int. J. Drug Dev. & Res., 11(1), 100–105.
  17. Singh, P., & Verma, R. (2020). Risk Assessment Tools in QbD: A Comprehensive Review. Int. J. Pharm. Sci. Invent., 9(6), 45–50.
  18. Joshi, R. D., & Mehta, T. A. (2018). A Review on Quality by Design (QbD). Int. J. Adv. Pharm., 7(1), 32–37.
  19. Desai, R. J., & Shah, D. R. (2022). Benefits and Challenges of QbD in the Indian Pharmaceutical Sector. Indian Drugs, 59(4), 12–18.
  20. Bhatt, D. A., & Rane, S. I. (2011). QbD: A Systematic Approach to Pharmaceutical Development. Pharma Times, 43(6), 16–20.
  21. Hinrichs WLJ, Möschwitzer JP. Quality by Design Approaches for Oral Drug Delivery Formulations. Adv Drug Deliv Rev. 2016;95:27–39.
  22. Raju NJ, et al. Challenges in Implementing QbD in Pharmaceutical Industries. Int J Pharm Sci Rev Res. 2014;28(1):72–78.
  23. Bolton S, Bon C. Pharmaceutical Statistics: Practical and Clinical Applications. 5th ed. CRC Press; 2009.
  24. Bhutani H, et al. Risk Assessment Tools in Pharmaceutical QbD. Pharm Anal Acta. 2014;5(6):1–8.
  25. Juran JM. Juran on Quality by Design: The New Steps for Planning Quality into Goods and Services. The Free Press; 1992.
  26. Narang AS, Desai D, Badawy S. Impact of Excipient Selection on Quality by Design (QbD) in Drug Product Development. AAPS PharmSciTech. 2012;13(3):1208–1217.
  27. Chow SC, Liu JP. Design and Analysis of Clinical Trials: Concepts and Methodologies. 3rd ed. Wiley; 2013.
  28. Xie W, Ji S, et al. Development of Controlled Release Systems Using QbD Approach. Eur J Pharm Biopharm. 2015;92:1–13.
  29. Kaur A, Kaur G. Application of QbD in Analytical Method Development: A Review. Int J Pharm Chem Biol Sci. 2013;3(4):1235–1244.
  30. Ferreiro I, et al. The Use of DoE in Pharmaceutical Development: An Industrial Perspective. Drug Dev Ind Pharm. 2020;46(7):1091–1103.

Photo
Rasheeda Parveen
Corresponding author

Roorkee College of Pharmacy, Dist. Haridwar, Uttarakhand.

Photo
Amit Chaudhary
Co-author

Roorkee College of Pharmacy, Dist. Haridwar, Uttarakhand.

Rasheeda Parveen*, Amit Chaudhary, Improvement in Designing, Formulation, and Development through QBD-Based SOPs: A Review, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 5, 3799-3809. https://doi.org/10.5281/zenodo.15489955

More related articles
Personalized Drug In Cancer Forestallment And Trea...
Prasanth Yerramsetti, Priyanka Gummadi, Abhishek Dey, ...
Improvement Of The Product Robustness Of Allopurin...
Pavithran P., Vasanthan A, Senthilkumar K. L. , ...
Role of Hospital Administrator in Medical and Clin...
Parth Kumar Pandey, Gopal, Shivshankar Tiwari, Ishani Debnath, Ma...
Related Articles
Nanoemulsion: An Emerging Approach For The Enhancement Of Solubility In Lipophil...
E. P. Punnya, Ranitha R., Megha P. M., Sreethu K. Sreedharan, ...
Assessment of Microbial Contamination of Coins Collected from Local Market of Kh...
Sujit Nagare, Sneha Limbani, Shrushti Chalke, Raj Ambre, Vipul Sansare, ...
Drug Effectiveness Through Clinical Drug Evaluation Recent Study ...
Tejas S. kapase , Yashraj Narake, Omkar Gongane, Sourabh patil, Sachin Navale, Nilesh Chougule, ...
Personalized Drug In Cancer Forestallment And Treatment As Well As Its Unborn Di...
Prasanth Yerramsetti, Priyanka Gummadi, Abhishek Dey, ...
More related articles
Personalized Drug In Cancer Forestallment And Treatment As Well As Its Unborn Di...
Prasanth Yerramsetti, Priyanka Gummadi, Abhishek Dey, ...
Improvement Of The Product Robustness Of Allopurinol With Minor Changes...
Pavithran P., Vasanthan A, Senthilkumar K. L. , ...
Role of Hospital Administrator in Medical and Clinical Operations in The Hospita...
Parth Kumar Pandey, Gopal, Shivshankar Tiwari, Ishani Debnath, Mansi Gupta, ...
Personalized Drug In Cancer Forestallment And Treatment As Well As Its Unborn Di...
Prasanth Yerramsetti, Priyanka Gummadi, Abhishek Dey, ...
Improvement Of The Product Robustness Of Allopurinol With Minor Changes...
Pavithran P., Vasanthan A, Senthilkumar K. L. , ...
Role of Hospital Administrator in Medical and Clinical Operations in The Hospita...
Parth Kumar Pandey, Gopal, Shivshankar Tiwari, Ishani Debnath, Mansi Gupta, ...