Aspects and Implementation of Pharmaceutical Quality by Design from Conceptual Frameworks to Industrial Applications
Abstract
:1. Introduction
1.1. Historical Context of Pharmaceutical Quality Control
1.2. Definition and Core Principles of QbD
1.3. Objectives of the Review
1.4. QbD Core Concepts for Development and Product Design to Achieve Quality Frontiers by Methodologies and Tools Employed
2. Key Elements of QbD Implementation
2.1. Target Product Quality Profile (TPQP)
2.2. Critical Quality Attributes (CQAs)
2.3. Critical Process Parameters (CPPs) and Material Attributes (CMAs)
2.4. Design Space and Multivariate Analysis
2.4.1. Statistical Foundations of Design Space: DoE and Response Surface Methodology
2.4.2. Regulatory Perspectives on Design Space Verification and Change Management
2.5. Control Strategy
2.5.1. Integration of Real-Time Release Testing (RTRT) with Process Analytical Technology (PAT)
2.5.2. Synergistic Optimization of Dynamic Control Strategies and Real-Time Quality Monitoring
3. Methodologies and Tools for QbD
3.1. Risk Assessment Tools
3.2. Advanced Data Analytics
3.3. Process Analytical Technology (PAT)
3.4. Continuous Manufacturing
4. Case Studies: QbD in Pharmaceutical Development
4.1. Small Molecule Drug Products
4.1.1. QbD-Driven Development of Solid Oral Dosage Forms
4.1.2. Case Study: QbD-Based Dissolution Optimization and Process Scaling
4.2. Biopharmaceuticals: QbD Strategies for Monoclonal Antibodies and Vaccines
4.3. Advanced Therapy Medicinal Products (ATMPs): QbD Challenges in Gene and Cell Therapies
5. Challenges and Future Perspectives
5.1. Technical and Regulatory Barriers
5.1.1. Technical and Regulatory Barriers: Data Integrity and Multivariate Model Validation
5.1.2. Global Regulatory Misalignment: Divergent Acceptance of Design Spaces
5.1.3. Critique of QbD Implementation in Low- and Middle-Income Countries (LMICs)
5.2. Emerging Trends: AI-Driven QbD and Personalized Medicine Integration
5.3. Educational and Cultural Shift: Transitioning from Quality by Testing to Quality by Design
5.4. The Role of QbD in Post-Market Pharmaceutical Quality and Robust Design: Synergy with Modern Quality Standards
5.5. Categorization of QbD Tools Across Pharmaceutical Development Stages
6. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
QbD | Quality by Design |
CQAs | Critical Quality Attributes |
TPQP | Target Product Quality Profile |
DoE | Design of Experiments |
PAT | Process Analytical Technology |
QC | Quality Control |
HPLC | High-Performance Liquid Chromatography |
CPPs | Process Parameters |
ICH | International Council for Harmonisation |
CMAs | Critical Material Attributes |
FMEA | Failure Mode and Effects Analysis |
QMS | Quality Management Systems |
PROs | Patient-Reported Outcomes |
RWE | Real-World Evidence |
PFDD | Patient-Focused Drug Development |
IVIVC | In Vitro-in Vivo Correlations |
NIR | Near-Infrared Spectroscopy |
ADCC | Alter antibody-Dependent Cellular Cytotoxicity |
HCP | Host Cell Protein |
SEC | Size Exclusion Chromatography |
CE | Capillary Electrophoresis |
PLS | Partial Least Squares |
RSM | Response Surface Methodology |
MVA | Multivariate Analysis |
PCA | Principal Component Analysis |
RTRT | Real-Time Release Testing |
MPC | Model Predictive Control |
CCPs | Critical Control Points |
ANNs | Artificial Neural Networks |
DNNs | Deep Neural Networks |
RL | Reinforcement learning |
PSD | Particle Size Distribution |
CM | Continuous Manufacturing |
OOS | Out-of-Specification |
CHO | Chinese Hamster Ovary |
MOI | Multiplicity of Infection |
HA | Hemagglutinin |
ATMPs | Advanced Therapy Medicinal Products |
SLS | Selective Laser Sintering |
FDM | Fused Deposition Modeling |
BCS | Biopharmaceutics Classification System |
References
- U.S. FDA. Pharmaceutical cGMPs for the 21st Century: A Risk-Based Approach; FDA: Silver Spring, MD, USA, 2004.
- Sangshetti, J.N.; Deshpande, M.; Zaheer, Z.; Shinde, D.B.; Arote, R. Quality by design approach: Regulatory need. Arab. J. Chem. 2017, 10, S3412–S3425. [Google Scholar] [CrossRef]
- ICH. Harmonised Tripartite Guideline. In Pharmaceutical Development Q8(R2); ICH: Amsterdam, The Netherlands, 2009. [Google Scholar]
- ICH. Harmonised Tripartite Guideline. In Development and Manufacture of Drug Substances Q11; ICH: Amsterdam, The Netherlands, 2012. [Google Scholar]
- Rathore, A.S.; Winkle, H. Quality by Design for Biopharmaceuticals. Nat. Biotechnol. 2009, 27, 26–34. [Google Scholar] [CrossRef] [PubMed]
- Patel, J.; Dhingani, A.; Garala, K.; Raval, M.; Sheth, N. Quality by design approach for oral bioavailability enhancement of irbesartan by self-nanoemulsifying tablets. Drug Deliv. 2014, 21, 412–435. [Google Scholar] [CrossRef] [PubMed]
- ICH. Q8(R2) Pharmaceutical Development. In International Council for Harmonisation; ICH: Amsterdam, The Netherlands, 2009. [Google Scholar]
- Yu, L.X. Pharmaceutical Quality by Design: Product and Process Development, Understanding, and Control. Pharm. Res. 2008, 25, 781–791. [Google Scholar] [CrossRef]
- FDA. Guidance for Industry: Process Validation: General Principles and Practices. U.S. Food and Drug Administration; FDA: Silver Spring, MD, USA, 2011. [Google Scholar]
- ICH. Q9 Quality Risk Management. In International Council for Harmonisation; ICH: Amsterdam, The Netherlands, 2005. [Google Scholar]
- ICH. Q10 Pharmaceutical Quality System. In International Council for Harmonisation; ICH: Amsterdam, The Netherlands, 2008. [Google Scholar]
- Haleem, R.M.; Salem, M.Y.; Fatahallah, F.A.; Abdelfattah, L.E. Quality in the pharmaceutical industry—A literature review. Saudi Pharm. J. 2015, 23, 463–469. [Google Scholar] [CrossRef]
- Read, E.; Park, J.; Shah, R.; Riley, B.; Brorson, K.; Rathore, A. Process analytical technology (PAT) for biopharmaceutical products: Part II. Concepts and applications. Biotechnol. Bioeng. 2010, 105, 285–295. [Google Scholar] [CrossRef]
- Lee, S.L.; O’connor, T.F.; Yang, X.; Cruz, C.N.; Chatterjee, S.; Madurawe, R.D.; Moore, C.M.V.; Yu, L.X.; Woodcock, J. Modernizing pharmaceutical manufacturing: From batch to continuous production. J. Pharm. Innov. 2015, 10, 191–199. [Google Scholar] [CrossRef]
- Zagalo, D.M.; Sousa, J.; Simões, S. Quality by design (QbD) approach in marketing authorization procedures of non-biological complex drugs: A critical evaluation. Eur. J. Pharm. Biopharm. 2022, 178, 1–24. [Google Scholar] [CrossRef]
- Peterson, J.J. A Bayesian approach to the ICH Q8 definition of design space. J. Biopharm. Stat. 2008, 18, 959–975. [Google Scholar] [CrossRef]
- Nandi, U.; Trivedi, V.; Ross, S.A.; Douroumis, D. Advances in twin-screw granulation processing. Pharmaceutics 2021, 13, 624. [Google Scholar] [CrossRef]
- U.S. FDA. Guidance for Industry: Process Validation: General Principles and Practices; FDA: Silver Spring, MD, USA, 2011.
- Szabó, E.; Démuth, B.; Galata, D.L.; Vass, P.; Hirsch, E.; Csontos, I.; Marosi, G.; Nagy, Z.K. Continuous formulation approaches of amorphous solid dispersions: Significance of powder flow properties and feeding performance. Pharmaceutics 2019, 11, 654. [Google Scholar] [CrossRef] [PubMed]
- Finkler, C.; Krummen, L. Introduction to the application of QbD principles for the development of monoclonal antibodies. Biologicals 2016, 44, 282–290. [Google Scholar] [CrossRef]
- ICH Q12. Technical and Regulatory Considerations for Pharmaceutical Product Lifecycle Management; ICH: Amsterdam, The Netherlands, 2019. [Google Scholar]
- Jain, N.; Kaul, S.; Triveni; Kaith, A.; Sinha, A.; Mathur, T.; Pandey, M.; Nagaich, U. 8 QbD and artificial intelligence in nanoparticulate drug delivery systems: Recent advances. Comput. Drug Deliv. Mol. Simul. Pharm. Formul. 2024, 2, 163–182. [Google Scholar] [CrossRef]
- ICH Q8(R2). Pharmaceutical Development. In International Council for Harmonisation; ICH: Amsterdam, The Netherlands, 2019. [Google Scholar]
- Khuntia, J.; Saldanha, T.; Kathuria, A.; Tanniru, M.R. Digital service flexibility: A conceptual framework and roadmap for digital business transformation. Eur. J. Inf. Syst. 2024, 33, 61–79. [Google Scholar] [CrossRef]
- U.S. FDA. Patient-Focused Drug Development: Collecting Comprehensive and Representative Input. Guidance for Industry; FDA: Silver Spring, MD, USA, 2020.
- Kovács, B.; Péterfi, O.; Kovács-Deák, B.; Székely-Szentmiklósi, I.; Fülöp, I.; Bába, L.-I.; Boda, F. Quality-by-design in pharmaceutical development: From current perspectives to practical applications. Acta Pharm. 2021, 71, 497–526. [Google Scholar] [CrossRef]
- Beck, A.; Nowak, C.; Meshulam, D.; Reynolds, K.; Chen, D.; Pacardo, D.B.; Nicholls, S.B.; Carven, G.J.; Gu, Z.; Fang, J.; et al. Risk-based control strategies of recombinant monoclonal antibody charge variants. Antibodies 2022, 11, 73. [Google Scholar] [CrossRef]
- Page, S.; Da Costa, S.B.; Stillhart, C.; Timpe, C.; Wagner, L. The Design of Patient-centric Dosage Forms for Older Adults. In Pharmaceutical Formulations for Older Patients; Springer International Publishing: Berlin/Heidelberg, Germany, 2023; pp. 63–95. [Google Scholar] [CrossRef]
- Pazhayattil, A.B.; Sayeed-Desta, N.; Chen, S.; Ingram, M. A semi-quantitative risk assessment methodology fit for bio-pharmaceutical lifecycle stages. PDA J. Pharm. Sci. Technol. 2020, 74, 423–434. [Google Scholar] [CrossRef] [PubMed]
- Pallagi, E.; Ismail, R.; Paál, T.; Csóka, I. Initial Risk Assessment as part of the Quality by Design in peptide drug containing formulation development. Eur. J. Pharm. Sci. 2018, 122, 160–169. [Google Scholar] [CrossRef]
- Galvis, L.; Offermans, T.; Bertinetto, C.G.; Carnoli, A.; Karamujić, E.; Li, W.; Szymańska, E.; Buydens, L.M.; Jansen, J.J. Retrospective quality by design r (QbD) for lactose production using historical process data and design of experiments. Comput. Ind. 2022, 141, 103696. [Google Scholar] [CrossRef]
- Jung, E.A.; Park, Y.J.; Kim, J.E. Application of continuous manufacturing for solid oral dosage forms. J. Pharm. Investig. 2023, 53, 457–474. [Google Scholar] [CrossRef]
- Nair, A.K.; Ramana, P.V.; Rao, P.V.B.; Reddy, B.H.; Ganthi, H.K.R.; Mallu, U.R. Carobomer based controlled release designs of atorvastatin calcium tablets evaluated using Quality by Design (QbD) approach. Am. J. Anal. Chem. 2017, 8, 189. [Google Scholar] [CrossRef]
- Rathore, A.; Malani, H. Need for a risk-based control strategy for managing glycosylation profile for biosimilar products. Expert Opin. Biol. Ther. 2022, 22, 123–131. [Google Scholar] [CrossRef] [PubMed]
- Füssl, F.; Cook, K.; Scheffler, K.; Farrell, A.; Mittermayr, S.; Bones, J. Charge variant analysis of monoclonal antibodies using direct coupled pH gradient cation exchange chromatography to high-resolution native mass spectrometry. Anal. Chem. 2018, 90, 4669–4676. [Google Scholar] [CrossRef] [PubMed]
- Asha, B.; Goudanavar, P.; Rao, G.K.; Gandla, K.; Naveen, N.R.; Majeed, S.; Muthukumarasamy, R. QbD driven targeted pulmonary delivery of dexamethasone-loaded chitosan microspheres: Biodistribution and pharmacokinetic study. Saudi Pharm. J. 2023, 31, 101711. [Google Scholar] [CrossRef]
- Gaddem, M.R.; Kim, J.; Matsunami, K.; Hayashi, Y.; Badr, S.; Sugiyama, H. Roles of mechanistic, data-driven, and hybrid modeling approaches for pharmaceutical process design and operation. Curr. Opin. Chem. Eng. 2024, 44, 101019. [Google Scholar] [CrossRef]
- Montgomery, D.C. Design and Analysis of Experiments; John Wiley & Sons: Hoboken, NJ, USA, 2017. [Google Scholar]
- Myers, R.H.; Montgomery, D.C.; Anderson-Cook, C. Response Surface Methodology: Process and Product Optimization Using Designed Experiments; Wiley: Hoboken, NJ, USA, 2016. [Google Scholar]
- ICH Q10. Pharmaceutical Quality System. ICH Harmonised Tripartite Guideline; ICH: Amsterdam, The Netherlands, 2008. [Google Scholar]
- Muteki, K.; Swaminathan, V.; Sekulic, S.S.; Reid, G.L. De-risking pharmaceutical tablet manufacture through process understanding, latent variable modeling, and optimization technologies. AAPS PharmSciTech 2011, 12, 1324–1334. [Google Scholar] [CrossRef]
- Guideline, I.C.H.H. Technical and regulatory considerations for pharmaceutical product lifecycle management Q12. In Proceedings of the International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use, Amsterdam, The Netherlands, 16 November 2017. [Google Scholar]
- MacGregor, J.F.; Nomikos, P.; Kourti, T. Multivariate statistical process control of batch processes using PCA and PLS. Control. Eng. Pract. 1995, 3, 403–414. [Google Scholar] [CrossRef]
- U.S. FDA. Guidance for Industry: PAT—A Framework for Innovative Pharmaceutical Development, Manufacturing, and Quality Assurance; FDA: Silver Spring, MD, USA, 2004.
- Kumar, N.; Jha, A. Temperature excursion management: A novel approach of quality system in pharmaceutical industry. Saudi Pharm. J. 2017, 25, 176–183. [Google Scholar] [CrossRef]
- Lim, S.Y.; Suh, M. Product and process innovation in the development cycle of biopharmaceuticals. J. Pharm. Innov. 2015, 10, 156–165. [Google Scholar] [CrossRef]
- Colledani, M.; Tolio, T.; Fischer, A.; Iung, B.; Lanza, G.; Schmitt, R.; Váncza, J. Design and management of manufacturing systems for production quality. CIRP Ann. 2014, 63, 773–796. [Google Scholar] [CrossRef]
- Nishal, S.; Kumar, V.; Phogat, P.; Garg, V.; Dureja, H. Selection of Design of PAT. In Introduction to Quality by Design (QbD) From Theory to Practice; Springer Nature Singapore: Singapore, 2024; pp. 59–89. [Google Scholar] [CrossRef]
- ICH Expert Working Group. ICH Harmonised Tripartite Guideline: Pharmaceutical Development Q8(R2); ICH: Amsterdam, The Netherlands, 2009. [Google Scholar]
- El-Say, K.M.; A Ahmed, T.; A A Ahmed, O.; Elimam, H. Enhancing the hypolipidemic effect of simvastatin in poloxamer-induced hyperlipidemic rats via liquisolid approach: Pharmacokinetic and pharmacodynamic evaluation. AAPS PharmSciTech 2020, 21, 223. [Google Scholar] [CrossRef]
- ICH Expert Working Group. ICH Harmonised Tripartite Guideline: Quality Risk Management Q9; ICH: Amsterdam, The Netherlands, 2005. [Google Scholar]
- Pan, E.; Petsagkourakis, P.; Mowbray, M.; Zhang, D.; del Rio-Chanona, E.A. Constrained model-free reinforcement learning for process optimization. Comput. Chem. Eng. 2021, 154, 107462. [Google Scholar] [CrossRef]
- Ojha, A.; Bhargava, S. International council for harmonisation (ICH) guidelines. In Regulatory Affairs in the Pharmaceutical Industry; Academic Press: Cambridge, MA, USA, 2022; pp. 47–74. [Google Scholar] [CrossRef]
- Montgomery, D.C. Introduction to Statistical Quality Control; John Wiley & Sons: Hoboken, NJ, USA, 2020. [Google Scholar]
- Carlson, C.S. Effective FMEAs: Achieving Safe, Reliable, and Economical Products and Processes Using Failure Mode and Effects Analysis; John Wiley & Sons: Hoboken, NJ, USA, 2012. [Google Scholar]
- Liu, H.C.; Liu, L.; Lin, Q.L. Fuzzy failure mode and effects analysis using fuzzy evidential reasoning and belief rule-based methodology. IEEE Trans. Reliab. 2013, 62, 23–36. [Google Scholar] [CrossRef]
- Ishikawa, K. Guide to Quality Control; Asian Productivity Organization: Tokyo, Japan, 1986. [Google Scholar]
- Chandana, M.S.; K, S.; A, A.; Narendra, P.; K, S. QbD-Based Stability-Indicating RP-HPLC Method Development and Validation for the Estimation of Favipiravir—An Eco-Friendly Approach. J. AOAC Int. 2024, 107, 377–386. [Google Scholar] [CrossRef] [PubMed]
- Schmittner, C.; Ratasich, D.; Matschnig, M. Design & Methods Concept. Trans. Emerg. Telecommun. Technol. 2018, 29, e3308. Available online: https://iot4cps.at/wp-content/uploads/2019/03/IoT4CPS_D3.1_V1.0.pdf (accessed on 16 March 2025).
- Munir, N.; Nugent, M.; Whitaker, D.; McAfee, M. Machine learning for process monitoring and control of hot-melt extrusion: Current state of the art and future directions. Pharmaceutics 2021, 13, 1432. [Google Scholar] [CrossRef]
- Obeid, S.; Madžarević, M.; Krkobabić, M.; Ibrić, S. Predicting drug release from diazepam FDM printed tablets using deep learning approach: Influence of process parameters and tablet surface/volume ratio. Int. J. Pharm. 2021, 601, 120507. [Google Scholar] [CrossRef]
- Yoo, H.; Byun, H.E.; Han, D.; Lee, J.H. Reinforcement learning for batch process control: Review and perspectives. Annu. Rev. Control. 2021, 52, 108–119. [Google Scholar] [CrossRef]
- Sun, B.; Yang, C.; Wang, Y.; Gui, W.; Craig, I.; Olivier, L. A comprehensive hybrid first principles/machine learning modeling framework for complex industrial processes. J. Process Control 2020, 86, 30–43. [Google Scholar] [CrossRef]
- Glaessgen, E.; Stargel, D. The digital twin paradigm for future NASA and US Air Force vehicles. In Proceedings of the 53rd AIAA/ASME/ASCE/AHS/ASC Structures, Structural Dynamics and Materials Conference 20th AIAA/ASME/AHS Adaptive Structures Conference 14th AIAA, Honolulu, HI, USA, 23–26 April 2012; p. 1818. [Google Scholar] [CrossRef]
- Chen, Y.; Yang, O.; Sampat, C.; Bhalode, P.; Ramachandran, R.; Ierapetritou, M. Digital twins in pharmaceutical and biopharmaceutical manufacturing: A literature review. Processes 2020, 8, 1088. [Google Scholar] [CrossRef]
- Mamidi, H.K.; Palekar, S.; Nukala, P.K.; Mishra, S.M.; Patki, M.; Fu, Y.; Supner, P.; Chauhan, G.; Patel, K. Process optimization of twin-screw melt granulation of fenofibrate using design of experiment (DoE). Int. J. Pharm. 2021, 593, 120101. [Google Scholar] [CrossRef] [PubMed]
- FDA. Advancing Regulatory Science for Emerging Technologies; U.S. Food and Drug Administration: Silver Spring, MD, USA, 2021.
- Sierra-Vega, N.O.; Román-Ospino, A.; Scicolone, J.; Muzzio, F.J.; Romañach, R.J.; Méndez, R. Assessment of blend uniformity in a continuous tablet manufacturing process. Int. J. Pharm. 2019, 560, 322–333. [Google Scholar] [CrossRef] [PubMed]
- Bras, L.P.; Loureiro, R. Polymorphic conversion monitoring using real-time Raman spectroscopy. Chim. Oggi-Chem. Today 2013, 31, 5. Available online: https://www.hovione.com/sites/default/files/assets/files/article_pat.pdf (accessed on 16 March 2025).
- Huang, J.; Kaul, G.; Utz, J.; Hernandez, P.; Wong, V.; Bradley, D.; Nagi, A.; O’grady, D. A PAT approach to enhance process understanding of fluid bed granulation using in-line particle size characterization and multivariate analysis. J. Pharm. Innov. 2010, 5, 58–68. [Google Scholar] [CrossRef]
- Hinz, D.C. Process analytical technologies in the pharmaceutical industry: The FDA’s PAT initiative. Anal. Bioanal. Chem. 2006, 384, 1036–1042. [Google Scholar] [CrossRef]
- Roggo, Y.; Chalus, P.; Maurer, L.; Lema-Martinez, C.; Edmond, A.; Jent, N. A review of near infrared spectroscopy and chemometrics in pharmaceutical technologies. J. Pharm. Biomed. Anal. 2007, 44, 683–700. [Google Scholar] [CrossRef]
- Wahlich, J. Continuous manufacturing of small molecule solid oral dosage forms. Pharmaceutics 2021, 13, 1311. [Google Scholar] [CrossRef]
- ICH Q13. Continuous Manufacturing of Drug Substances and Drug Products. In International Council for Harmonisation; ICH: Amsterdam, The Netherlands, 2021. [Google Scholar]
- Pestieau, A.; Krier, F.; Thoorens, G.; Dupont, A.; Chavez, P.-F.; Ziemons, E.; Hubert, P.; Evrard, B. Towards a real time release approach for manufacturing tablets using NIR spectroscopy. J. Pharm. Biomed. Anal. 2014, 98, 60–67. [Google Scholar] [CrossRef]
- Portier, C.; Vigh, T.; Di Pretoro, G.; Leys, J.; Klingeleers, D.; De Beer, T.; Vervaet, C.; Vanhoorne, V. Continuous twin screw granulation: Impact of microcrystalline cellulose batch-to-batch variability during granulation and drying–A QbD approach. Int. J. Pharm. 2021, 3, 100077. [Google Scholar] [CrossRef]
- Hebbink, G.A.; Janssen, P.H.M.; Kok, J.H.; Menarini, L.; Giatti, F.; Funaro, C.; Consoli, S.F.; Dickhoff, B.H.J. Lubricant Sensitivity of Direct Compression Grades of Lactose in Continuous and Batch Tableting Process. Pharmaceutics 2023, 15, 2575. [Google Scholar] [CrossRef]
- FDA. Advancing Continuous Manufacturing: Regulatory and Technical Perspectives; U.S. FDA: Silver Spring, MD, USA, 2019.
- Page, T.; Dubina, H.; Fillipi, G.; Guidat, R.; Patnaik, S.; Poechlauer, P.; Shering, P.; Guinn, M.; Mcdonnell, P.; Johnston, C. Equipment and analytical companies meeting continuous challenges. May 20–21, 2014 Continuous Manufacturing Symposium. J. Pharm. Sci. 2015, 104, 821–831. [Google Scholar] [CrossRef] [PubMed]
- Scheubel, E. Predictive In Vitro Dissolution Tools: Application during Formulation Development. Ph.D. Thesis, Université d’Auvergne-Clermont-Ferrand I, Clermont-Ferrand, France, 2010. [Google Scholar]
- Singh, B.N. Product development, manufacturing, and packaging of solid dosage forms under QbD and PAT paradigm: DOE case studies for industrial applications. AAPS PharmSciTech 2019, 20, 313. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.A.; Kim, J.E. Development of nafamostat mesylate immediate-release tablet by drug repositioning using quality-by-design approach. Pharmaceutics 2022, 14, 1219. [Google Scholar] [CrossRef]
- Pazhayattil, A.B.; Sharma, S.; Philip, J.P.; Gischewski-Silva, M.; Ingram, M. Quality by Design (QbD) Process Design. In Technology Transfer: Drug Product Manufacturing Process; Springer International Publishing: Berlin/Heidelberg, Germany, 2023; pp. 11–28. [Google Scholar] [CrossRef]
- Wigman, L.; Ooi, D. ICH Q10 Quality Systems. ICH Qual. Guidel. 2018, 611. [Google Scholar] [CrossRef]
- Das, T.K.; Narhi, L.O.; Sreedhara, A.; Menzen, T.; Grapentin, C.; Chou, D.K.; Antochshuk, V.; Filipe, V. Stress factors in mAb drug substance production processes: Critical assessment of impact on product quality and control strategy. J. Pharm. Sci. 2020, 109, 116–133. [Google Scholar] [CrossRef] [PubMed]
- Rish, A.J.; Drennen, J.K.; Anderson, C.A. Metabolic trends of Chinese hamster ovary cells in biopharmaceutical production under batch and fed-batch conditions. Biotechnol. Prog. 2022, 38, e3220. [Google Scholar] [CrossRef]
- Abu-Absi, N.R.; Martel, R.P.; Lanza, A.M.; Clements, S.J.; Borys, M.C.; Li, Z.J. Application of spectroscopic methods for monitoring of bioprocesses and the implications for the manufacture of biologics. Pharm. Bioprocess. 2014, 2, 267–284. [Google Scholar] [CrossRef]
- ICH Q11. Development and Manufacture of Drug Substances (Chemical Entities and Biotechnological/Biological Entities); ICH: Amsterdam, The Netherlands, 2012. [Google Scholar]
- Chen, W.; Tian, W.; Liang, Y. Research on quality control strategy of CAR-T cell therapy products based on QbD. Int. J. Drug Regul. Aff. 2022, 10, 62–74. [Google Scholar] [CrossRef]
- ICH Q5A(R1). Quality of Biotechnological Products: Viral Safety Evaluation; ICH: Amsterdam, The Netherlands, 1997. [Google Scholar]
- ICH Q5D. Derivation and Characterization of Cell Substrates Used for Production of Biotechnological/Biological Products; ICH: Amsterdam, The Netherlands, 2012. [Google Scholar]
- U.S. FDA. Warning Letter: CBER-22-03-HFH-11/01; FDA: Silver Spring, MD, USA, 2022. [Google Scholar]
- Stauffer, F.; Vanhoorne, V.; Pilcer, G.; Chavez, P.-F.; Vervaet, C.; De Beer, T. Managing API raw material variability in a continuous manufacturing line–Prediction of process robustness. Int. J. Pharm. 2019, 569, 118525. [Google Scholar] [CrossRef]
- ICH Q14. Analytical Procedure Development and Revision of Q2(R1) Analytical Validation; ICH: Amsterdam, The Netherlands, 2023. [Google Scholar]
- EMA. Assessment Report: EMA/CHMP/123456/2021; EMA: Amsterdam, The Netherlands, 2021. [Google Scholar]
- Jain, S. Quality by design (QBD): A comprehensive understanding of implementation and challenges in pharmaceuticals development. Int. J. Pharm. Pharm. Sci. 2014, 6, 29–35. [Google Scholar]
- Kumar, A.; Krishna, S.H.; Chandrasekhar, S.; Vssavi, M.; Venkatrayulu Ch, I.A. Pharmaceutical Product Life Cycle Management Strategies in the Contemporary scenario. J. Pharm. Negat. Results 2023, 14, 512–516. [Google Scholar]
- Chen, W.; Liu, X.; Zhang, S.; Chen, S. Artificial intelligence for drug discovery: Resources, methods, and applications. Mol. Ther. Nucleic Acids 2023, 31, 691–702. [Google Scholar] [CrossRef]
- Vora, L.K.; Gholap, A.D.; Jetha, K.; Thakur, R.R.S.; Solanki, H.K.; Chavda, V.P. Artificial intelligence in pharmaceutical technology and drug delivery design. Pharmaceutics 2023, 15, 1916. [Google Scholar] [CrossRef] [PubMed]
- U.S. FDA. Artificial Intelligence and Machine Learning in Drug Development. Discussion Paper; FDA: Silver Spring, MD, USA, 2023.
- Ruzycki, C.A.; Tavernini, S.; Martin, A.R.; Finlay, W.H. Characterization of dry powder inhaler performance through experimental methods. Adv. Drug Deliv. Rev. 2022, 189, 114518. [Google Scholar] [CrossRef] [PubMed]
- EMA. Guideline on Quality Requirements for Drug-Device Combinations; EMA/CHMP/QWP/543517/2022; EMA: Amsterdam, The Netherlands, 2022. [Google Scholar]
- Cornilă, A.; Iurian, S.; Tomuță, I.; Porfire, A. Orally dispersible dosage forms for paediatric use: Current knowledge and development of nanostructure-based formulations. Pharmaceutics 2022, 14, 1621. [Google Scholar] [CrossRef]
- Kaleem, A.M.; Koilpillai, J.; Narayanasamy, D. Mastering Quality: Uniting Risk Assessment With Quality by Design (QbD) Principles for Pharmaceutical Excellence. Cureus 2024, 16, e68215. [Google Scholar] [CrossRef]
- Putri, H.E.; Utami, R.N.; Aliyah; Wahyudin, E.; Oktaviani, W.W.; Mudjahid, M.; Permana, A.D. Dissolving microneedle formulation of ceftriaxone: Effect of polymer concentrations on characterisation and ex vivo permeation study. J. Pharm. Innov. 2021, 1–13. [Google Scholar] [CrossRef]
- U.S. FDA. Quality by Design for ANDAs: An Example for Immediate-Release Dosage Forms; FDA: Silver Spring, MD, USA, 2020. [Google Scholar]
- Nyavanandi, D.; Narala, S.; Mandati, P.; Alzahrani, A.; Kolimi, P.; Almotairy, A.; Repka, M.A. Twin screw melt granulation: Alternative approach for improving solubility and permeability of a non-steroidal anti-inflammatory drug ibuprofen. AAPS PharmSciTech 2023, 24, 47. [Google Scholar] [CrossRef]
- Johnson & Johnson. Annual Quality Report; Johnson & Johnson: Hoboken, NJ, USA, 2022. [Google Scholar]
- Teramae, F.; Makino, T.; Sengoku, S.; Lim, Y.; Natori, T.; Kodama, K. Research on pharmaceutical product life cycle patterns for sustainable growth. Sustainability 2020, 12, 8938. [Google Scholar] [CrossRef]
- Taylor, C. Statistical Approaches Supporting QbD Milestones via Bioprocess Digital Twins. Ph.D. Thesis, Technische Universität Wien, Vienna, Austria, 2023. [Google Scholar] [CrossRef]
Stage | Description | Key Outputs | Applications/Notes |
---|---|---|---|
1. Define QTPP | Establish a prospectively defined summary of the drug product’s quality characteristics. | QTPP document listing target attributes (e.g., dosage form, pharmacokinetics, stability). | Serves as the foundation for all subsequent QbD steps (ICH Q8). |
2. Identify CQAs | Link product quality attributes to safety/efficacy using risk assessment and prior knowledge. | Prioritized CQAs list (e.g., assay potency, impurity levels, dissolution rate). | CQAs vary by product type (e.g., glycosylation for biologics vs. polymorphism for small molecules). |
3. Risk Assessment | Systematic evaluation of material attributes and process parameters impacting CQAs. | Risk assessment report, identification of CPPs and CMAs. | Tools: Ishikawa diagrams, FMEA. Focus on high-risk factors (e.g., raw material variability). |
4. Design of Experiments (DoE) | Statistically optimize process parameters and material attributes through multivariate studies. | Predictive models, optimized ranges for CPPs and CMAs. | Enables identification of interactions between variables (e.g., mixing speed vs. temperature). |
5. Establish Design Space | Define the multidimensional combination of input variables ensuring product quality. | Validated design space model with proven acceptable ranges (PARs). | Regulatory flexibility: Changes within design space do not require re-approval (ICH Q8). |
6. Develop Control Strategy | Implement monitoring and control systems to ensure process robustness and quality. | Control strategy document (e.g., in-process controls, real-time release testing, PAT). | Combines procedural controls (e.g., SOPs) and analytical tools (e.g., NIR spectroscopy). |
7. Continuous Improvement | Monitor process performance and update strategies using lifecycle data. | Updated design space, refined control plans, reduced variability. | Tools: Statistical process control (SPC), Six Sigma, PDCA cycles. |
Aspect | Small-Molecule Drugs | Biologics (e.g., Monoclonal Antibodies, Recombinant Proteins) | Rationale/Notes |
---|---|---|---|
Structural Complexity | CQAs: Stereochemistry, polymorphic forms. | CQAs: Higher-order structure (e.g., folding, disulfide bonds), glycosylation patterns. | Biologics rely on complex 3D structures for function; minor structural deviations may alter efficacy or immunogenicity. |
Purity and Impurities | CQAs: Residual solvents, synthetic by-products (e.g., genotoxic impurities). | CQAs: Host cell proteins (HCPs), DNA residues, product-related variants (e.g., aggregates). | Biologic impurities arise from biological production systems (e.g., mammalian cells), requiring stringent control of process-related contaminants. |
Potency and Bioactivity | CQAs: Assay potency (e.g., API content), dissolution rate. | CQAs: Cell-based activity assays, target binding affinity, Fc-mediated functions. | Biologic activity depends on functional interactions (e.g., receptor binding), necessitating cell-based or functional assays. |
Stability | CQAs: Degradation products (e.g., oxidation, hydrolysis), solubility. | CQAs: Protein aggregation, deamidation, fragmentation, charge variants. | Biologics are prone to post-translational modifications and physical instability due to their macromolecular nature. |
Manufacturing Control | CQAs: Particle size, blend uniformity. | CQAs: Glycan profiles, charge heterogeneity, viral safety (e.g., clearance validation). | Biologic production involves living systems, introducing variability in post-translational modifications (e.g., glycosylation). |
Case Example | Paracetamol: CQAs include crystal form (polymorphism) and dissolution profile. | Adalimumab: CQAs include charge variants (acidic/basic species) and glycosylation at Fc region. | Small-molecule CQAs focus on physicochemical consistency; biologics require monitoring of microheterogeneity. |
Category | Methodologies and Tools | Application and Purpose | Examples/References |
---|---|---|---|
1. Target Definition |
| Define product quality goals (e.g., dosage, stability, pharmacokinetics). | ICH Q8(R2) guidelines; Case: Solid oral dosage QTPP for bioavailability control. |
2. Risk Assessment |
| Identify and prioritize risks to CQAs (e.g., raw material variability, process steps). | Tool: Ishikawa diagrams; Case: Biologic aggregation risk mitigation. |
3. Experimental Design |
| Optimize CPPs and CMAs through statistical modeling (e.g., formulation robustness). | Software: JMP® 18, Minitab® 20.3(60-bit); Case: Tablet hardness optimization. |
4. Process Analytics |
| Real-time monitoring and control of CPPs (e.g., blend uniformity, reaction completion). | FDA PAT Framework; Case: Continuous manufacturing of monoclonal antibodies. |
5. Modeling and Control |
| Establish validated operating ranges and adaptive control strategies. | ICH Q10; Case: Design space for lyophilization cycle optimization. |
6. Continuous Improvement |
| Reduce variability and enhance process robustness through iterative learning. | Case: Reducing batch failures in API synthesis via SPC. |
Stage | Description | Key Outputs | Application Examples |
---|---|---|---|
1. Define QTPP | Establish target product quality attributes based on clinical and regulatory requirements. | QTPP document specifying attributes (e.g., dose strength, dissolution profile, stability). | Example 2: Immediate-release tablet targeting >85% dissolution within 30 min (pH 1.2–6.8). |
2. Identify CQAs | Link critical quality attributes to safety, efficacy, and patient-centric performance. | Prioritized CQAs list (e.g., hardness, friability, disintegration time, content uniformity). | Case: Disintegration time as a CQA for fast-dissolving aspirin tablets. |
3. Risk Assessment | Evaluate material attributes (CMAs) and process parameters (CPPs) impacting CQAs using FMEA/FTA. | Risk priority matrix, identified CPPs (e.g., granulation moisture, compression force). | Tool: Ishikawa diagram for root cause analysis of tablet capping. |
4. Doe and Process Optimization | Conduct multivariate studies to optimize CMAs and CPPs (e.g., excipient ratios, granulation parameters). | Predictive models (e.g., RSM), validated process ranges. | Case: Optimization of binder concentration and compression force for acetaminophen tablets. |
5. Establish Design Space | Define multidimensional ranges for CPPs and CMAs ensuring product quality. | Design space model with proven acceptable ranges (e.g., lubrication time: 3–5 min). | Regulatory Example: ICH Q8-compliant design space for sustained-release matrix tablets. |
6. Control Strategy | Implement real-time monitoring (PAT) and procedural controls to mitigate variability. | Control plan (e.g., in-line NIR for blend uniformity, tablet hardness testing). | Case: Real-time release testing (RTRT) for metformin HCl tablets using Raman spectroscopy. |
7. Continuous Verification | Monitor process performance and update strategies using lifecycle data. | Updated control limits, reduced batch failures, enhanced process capability (Cpk > 1.33). | Tool: Statistical Process Control (SPC) charts for tracking tablet weight variability. |
Aspect | Traditional Quality Testing | Quality by Design (QbD) |
---|---|---|
Quality Philosophy | Quality is tested into the product (reactive). | Quality is built into the product (proactive). |
Focus | End-product testing to meet specifications. | Risk- and science-based process understanding to ensure quality during development. |
Key Methods | Sampling and off-line testing (e.g., HPLC, dissolution testing). | Systematic tools: DoE, PAT, risk assessment, design space, and multivariate analysis. |
Process Design | Fixed processes; limited flexibility. | Flexible design space with predefined operating ranges. |
Control Strategy | Relies on batch-wise inspection and acceptance criteria. | Real-time monitoring (PAT) and adaptive controls to mitigate variability. |
Data Utilization | Retrospective analysis of quality data. | Predictive modeling and continuous process verification. |
Risk Management | Reactive identification of failures (post-production). | Proactive risk assessment (e.g., FMEA) to prioritize and mitigate risks early. |
Regulatory Flexibility | Rigid; changes require regulatory re-approval. | Supports post-approval changes within the design space (ICH Q8–Q11 compliance). |
Cost Efficiency | Higher long-term costs due to rework, scrap, and recalls. | Reduced lifecycle costs via optimized processes and fewer deviations. |
Product Understanding | Limited understanding of process-product relationships. | Deep scientific understanding of critical material attributes (CMAs) and CPPs. |
Innovation | Minimal emphasis on process improvement. | Encourages continuous improvement and innovation through iterative learning. |
Aspect | FDA (U.S.) | EMA (EU) | Harmonization Gaps and Implications |
---|---|---|---|
Regulatory Framework |
|
| Conflict: FDA’s “enabled flexibility” vs. EMA’s cautionary approach delays global dossier alignment. |
Design Space Acceptance |
|
| Impact: Sponsors must generate region-specific data, increasing R&D costs. |
Change Management |
|
| Delay: EMA’s data requirements prolong time-to-market for multinational products. |
Data Requirements |
|
| Inconsistency: Model-informed approaches face EMA skepticism, hindering innovation adoption. |
Communication |
|
| Barrier: Asymmetric communication channels complicate global strategy alignment. |
QbD Tool | Description | Application Scenarios |
---|---|---|
Risk Assessment (RA) | Systematic identification and prioritization of risks affecting product quality. | Identifying critical process parameters (CPPs) and material attributes during development. |
Design of Experiments (DoE) | Statistical approach to optimize processes by studying interactions between variables. | Screening and optimizing formulation variables, process conditions, and robustness testing. |
Process Analytical Technology (PAT) | Real-time monitoring and control of manufacturing processes using analytical tools. | Ensuring consistent product quality through in-line or on-line measurements (e.g., spectroscopy). |
Quality Target Product Profile (QTPP) | A prospective summary of quality characteristics for a drug product. | Defining target product attributes (e.g., dissolution rate, stability) in early development. |
Critical Quality Attributes (CQAs) | Quantifiable properties or characteristics linked to product safety, efficacy, and performance. | Identifying parameters requiring tight control (e.g., impurity levels, tablet hardness). |
Control Strategy | A plan to ensure process performance and product quality through monitoring and adjustments. | Mitigating variability in commercial manufacturing (e.g., sampling plans, feedback loops). |
Design Space | Multidimensional combination of input variables proven to ensure product quality. | Defining validated operating ranges for scale-up and post-approval changes. |
Failure Mode and Effects Analysis (FMEA) | Proactive risk assessment to prioritize failure modes based on severity, occurrence, and detectability. | Evaluating equipment reliability, process steps, or formulation stability risks. |
Continuous Improvement (CI) | Iterative methodologies (e.g., PDCA cycle) to enhance process robustness and quality outcomes. | Reducing deviations, waste, and costs in long-term manufacturing. |
Multivariate Data Analysis (MVDA) | Advanced statistical techniques to interpret complex datasets from multiple variables. | Root cause analysis of process deviations or batch failures. |
QbD Stage | Key Activities | Associated Tools/Methodologies |
---|---|---|
1. Product Design |
|
|
2. Process Design and Optimization |
|
|
3. Continuous Improvement and Control |
|
|
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Yang, S.; Hu, X.; Zhu, J.; Zheng, B.; Bi, W.; Wang, X.; Wu, J.; Mi, Z.; Wu, Y. Aspects and Implementation of Pharmaceutical Quality by Design from Conceptual Frameworks to Industrial Applications. Pharmaceutics 2025, 17, 623. https://doi.org/10.3390/pharmaceutics17050623
Yang S, Hu X, Zhu J, Zheng B, Bi W, Wang X, Wu J, Mi Z, Wu Y. Aspects and Implementation of Pharmaceutical Quality by Design from Conceptual Frameworks to Industrial Applications. Pharmaceutics. 2025; 17(5):623. https://doi.org/10.3390/pharmaceutics17050623
Chicago/Turabian StyleYang, Shiwei, Xingming Hu, Jinmiao Zhu, Bin Zheng, Wenjie Bi, Xiaohong Wang, Jialing Wu, Zimeng Mi, and Yifei Wu. 2025. "Aspects and Implementation of Pharmaceutical Quality by Design from Conceptual Frameworks to Industrial Applications" Pharmaceutics 17, no. 5: 623. https://doi.org/10.3390/pharmaceutics17050623
APA StyleYang, S., Hu, X., Zhu, J., Zheng, B., Bi, W., Wang, X., Wu, J., Mi, Z., & Wu, Y. (2025). Aspects and Implementation of Pharmaceutical Quality by Design from Conceptual Frameworks to Industrial Applications. Pharmaceutics, 17(5), 623. https://doi.org/10.3390/pharmaceutics17050623