Shri Gajanan Maharaj Shikshan Prasarak Mandal’s Dnyanvilas College of pharmacy, Dudulgaon, Pune 411081
Quality by Design is a systematic approach used in pharmaceutical development that builds product quality into the formulation and manufacturing processes from the outset. Although its adoption is increasing within the generic drug industry, many professionals are still not deeply familiar with its principles or terminology. This paper outlines the core concepts of QbD, showing how it ensures reliable product quality by defining the desired product profile, determining the critical quality attributes, setting an appropriate design space, establishing a comprehensive control strategy, and overseeing the product throughout its lifecycle. By thoroughly evaluating how formulation ingredients and processing parameters influence product performance, QbD streamlines development, provides greater regulatory flexibility, and strengthens product consistency. Effective application of QbD requires strong collaboration among R&D, manufacturing, quality, and regulatory teams, aligned with international guidelines such as ICH Q8, Q9, and Q10. [1]
Since the FDA revised its current Good Manufacturing Practices (cGMP) in 2002, the pharmaceutical industry has increasingly recognized the importance of strengthening product quality. Traditional reliance on end-product testing alone has shown to be inefficient, slow, and reactive. This has encouraged the adoption of more forward-looking and structured quality approaches. As a result, Quality by Design (QbD) has gained prominence as a more dependable and effective strategy. Modern drug development emphasizes deliberately designing products that consistently meet quality expectations and establishing manufacturing processes that can reliably reproduce those outcomes. The International Council for Harmonisation (ICH) promotes a lifecycle-based, science-driven, and risk-focused model to ensure quality, with the overarching goal of delivering safe, high-quality medications to patients. [5]
ICH guideline Q8, which focuses on formulation and process development, underscores the need to build quality into the product from the earliest development phases rather than relying solely on final checks. This philosophy centers on selecting appropriate formulations and process conditions from the beginning, followed by continuous refinement throughout development. Its core concepts include comprehensive quality management and ongoing improvement. The QbD framework supports these principles by fostering a detailed understanding of both the product and the manufacturing process. For instance, if a batch does not meet its quality criteria because of raw-material variability, QbD calls for identifying the cause and extent of the variation, evaluating its effect, and applying monitoring or control mechanisms—such as real-time analytical tools—to maintain consistent quality. In contrast to traditional methods that simply tighten ranges or remove suspect materials, QbD provides a more holistic, efficient, and dependable path to quality assurance. [5]
Regulatory guidelines also provide direction on implementing QbD. As per ICH Q8 (R1), QbD is described as a “systematic development approach that begins with clearly defined objectives, emphasizes thorough understanding of both the product and the manufacturing process, and applies scientifically justified process controls supported by quality risk management.” Likewise, the FDA’s Process Analytical Technology (PAT) framework defines QbD as a methodology that utilizes real-time data to design, monitor, and control manufacturing operations, ensuring that the final product is safe and effective. The European Medicines Agency (EMA) and other international regulatory authorities have also incorporated QbD principles into their expectations for drug development. In essence, QbD offers a modern, science-based approach that enhances development efficiency, minimizes manual interventions through automation, and allows faster identification and resolution of quality-related issues. Instead of relying only on end-product testing, it emphasizes early detection of process deviations and timely corrective actions. [5]
QbD is not limited to product formulation and manufacturing; its principles can also be applied to the development of analytical methods, where it is referred to as Analytical Quality by Design (AQbD). This article highlights the value of incorporating QbD concepts into analytical processes, compares conventional analytical method development with the AQbD approach, and examines the regulatory framework supporting AQbD. It further describes how QbD elements can be implemented within analytical workflows and provides examples of successful application. Maxine K. Fritz from NSF Becker & Associates Consulting emphasizes that quality should be integrated from the outset, pointing out that while this may require an initial investment, neglecting to establish robust quality systems can result in much greater financial and operational challenges in the long run. [5]
A survey of the scientific literature indicates that the pharmaceutical industry has not yet fully grasped the concept of AQbD. Many professionals mistakenly equate it with general QbD, although AQbD focuses specifically on analytical method development. Furthermore, practical resources for implementing AQbD are limited, and knowledge of key tools like Design of Experiments (DoE) remains inadequate among analysts. This underscores the need for comprehensive clarification of AQbD principles, step-by-step guidance for laboratory application, and illustrative examples demonstrating its effectiveness in creating robust, scientifically sound analytical methods. [5]
Analytical Quality by Design applies QbD principles to the development, optimization, and lifecycle oversight of analytical procedures. The central idea is to embed quality into the method from the very beginning instead of confirming it only after development. Rather than depending on trial-and-error approaches, AQbD employs a structured, scientifically grounded process that includes risk assessment, planned experimental studies, and continuous refinement. [5]
Principle:
Analytical Quality by Design applies the principles of QbD to the creation and refinement of analytical methods. Rather than relying primarily on testing the final product, AQbD focuses on incorporating quality into the analytical method from the outset. Its objective is to develop methods that are scientifically robust, dependable, and consistently deliver accurate results. AQbD provides a structured approach for the systematic design, optimization, validation, and ongoing lifecycle management of analytical techniques. [3]
Analytical Quality through Design According to Fundamentals and Design Principle:
Analytical Quality through Design is grounded in the core QbD principles, which focus on clearly establishing how product attributes relate to the process parameters that influence them. QbD involves creating a multidimensional “design space” that illustrates how these factors interact. The same concepts used in developing manufacturing processes can be applied to designing analytical methods. [18]
In Analytical Quality by Design, the workflow begins with establishing the Analytical Target Profile (ATP), which defines the objective of the analysis and the expected performance criteria for the method. A comprehensive understanding of the analytical procedure is then achieved by identifying the Critical Method Parameters (CMPs) through detailed risk assessments and multivariate analysis techniques. The Method Operating Range, also referred to as the design space, determines the permissible limits for these CMPs, ensuring that the method reliably meets the designated Critical Method Attributes (CMAs). [18]
Analytical Target Profile:
Within the AQbD framework, the Analytical Target Profile (ATP) forms the cornerstone of analytical method development, similar to how the Quality Target Product Profile (QTPP) directs product design. It defines the purpose of the analytical procedure and sets quantifiable performance goals. The ATP also specifies the analyte of interest, the chosen analytical technique (such as HPLC, GC, ion chromatography, or HPTLC), and the essential characteristics the method must evaluate, including impurity detection and other critical analytical parameters. [12]
Critical Quality Attributes (CQAs):
Once the ATP is established, the next stage is to identify the Critical Quality Attributes. These refer to the physical, chemical, or biological properties that must be maintained within acceptable limits to ensure that the analytical method provides accurate and consistent results. CQAs are directly connected to method performance and play a vital role in achieving the desired analytical quality. [14]
Risk Assessment:
In AQbD, risk assessment examines how changes in analytical conditions—such as operator handling, instrument settings, sample characteristics, and environmental factors—can impact Critical Quality Attributes (CQAs). Following ICH Q9 guidelines, this process involves identifying potential risks, analyzing them, and evaluating their significance. Tools like flowcharts, checklists, and Ishikawa (fishbone) diagrams help categorize variables into major risks, noise factors, and experimental elements. More formalized approaches, including FMEA, FMECA, REM, HAZOP, and HACCP, enable semi-quantitative risk ranking. For example, FMEA assigns numerical scores based on severity, likelihood, and detectability, while REM categorizes risks as low, medium, or high according to their impact and frequency. [13]
Method Operational Domain (MOD):
The goal of the analyst is to define the method’s operational domain using data generated from Design of Experiments (DoE), often supported by response surface modeling. This domain represents the multidimensional range within which all Critical Method Attributes (CMAs) satisfy predefined acceptance limits, ensuring the method performs consistently and reliably. It is commonly illustrated using overlapping contour plots for various responses. A related concept, the Method Operable Design Region (MODR), outlines the specific conditions under which the analytical method consistently operates. For example, in developing an HPLC method, initial screening experiments are used to pinpoint important factors like the mobile phase ratio, column temperature, and flow rate. These are then refined and optimized through response surface methodology or similar optimization studies. [16]
Control Strategy:
The control strategy is developed using knowledge gained from the Method Operable Design Region and defines the actions required to maintain consistent method performance throughout its lifecycle. It is a dynamic approach that evolves during method development, validation, and routine use. While QbD based control strategies share similarities with conventional approaches, they place greater emphasis on statistical evaluation and a thorough understanding of the design spaceFor instance, factors such as mobile phase ratio, flow rate, detection wavelength, and column temperature—along with their influence on responses like peak area, retention time, and plate efficiency—can be evaluated together to develop a thorough and dependable control strategy. [16]
Regulatory Considerations for Analytical QbD:
Adopting Analytical Quality by Design (AQbD) enables pharmaceutical companies to enhance the reliability and consistency of analytical methods while minimizing regulatory challenges. The regulatory groundwork for AQbD dates back to August 2002, when the U.S. FDA launched the initiative “Pharmaceutical cGMPs for the 21st Century: A Risk-Based Approach,” highlighting the implementation of QbD principles and encouraging continuous improvement in manufacturing processes. By 2005, the FDA expected Chemistry, Manufacturing, and Controls (CMC) submissions to align with a QbD-oriented framework. These requirements were further supported by international guidelines, including ICH Q8 (R1) on pharmaceutical development, ICH Q9 on quality risk management, and ICH Q10 on the pharmaceutical quality system. While ICH Q8 (R2) does not explicitly cover analytical design space, its concepts—such as establishing design space, applying risk-based approaches, and pursuing continuous improvement—are widely applied in analytical method development to enhance method robustness (citation needed). [7][19]
In recent years, analytical method failures have become more frequent, particularly during method transfer and within quality control laboratories. This is partly due to limited robustness testing under ICH Q2, which provides flexibility during method validation. The rise in FDA warning letters related to quality control highlights the need for more reliable and robust analytical methods. As a result, integrating QbD principles into analytical method development is increasingly recognized as critical. According to Q10 guidance, analytical methods are a key component of the overall control strategy to ensure consistent product quality. [7][19]
Analytical Quality by Design: Tools
Analytical Quality by Design applies the QbD principles to analytical method development, facilitating the creation of methods that are reliable, robust, and thoroughly characterized. Unlike traditional approaches, AQbD is systematic, science-driven, and focused on risk management. A range of tools is utilized throughout the AQbD process to develop, optimize, validate, and maintain analytical methods. These tools support a deeper understanding of method performance, help identify critical factors, and guide the implementation of control strategies to maintain consistent quality throughout the method’s lifecycle. [15]
The Analytical Objective Profile (AOP) serves as the cornerstone of AQbD by defining the purpose of an analytical method and the quality criteria it must fulfill. It addresses the question: “What is the intended goal of the method?” Essential performance attributes outlined in the AOP include precision, accuracy, specificity (LOD/LOQ), linearity, range, and robustness. Defining the AOP at the beginning of method development establishes clear targets, ensuring the method is fit for its intended use and meets regulatory requirements. It also provides guidance for subsequent activities such as risk assessment, method validation, and lifecycle management. [15]
Risk assessment helps identify and prioritize factors that could impact method performance, allowing resources to be focused on the most influential variables. Commonly used tools include:
These approaches reduce unnecessary experimentation and focus on factors that have a meaningful impact on method performance. [15]
3. Design of Experiments (DoE):
DoE is a key statistical tool in AQbD that studies multiple variables simultaneously, providing a systematic understanding of method behavior. Benefits include identifying critical factors, understanding interactions, optimizing conditions for accuracy and precision, and building predictive models. Typical designs include:
DoE supports defining the Method Operable Design Region and ensures the method consistently meets ATP requirements. [15]
The MODR represents the multidimensional combination of Critical Method Parameters (CMPs) in which the analytical procedure consistently achieves the objectives defined in the Analytical Target Profile (ATP). Essential aspects include:
MODR is established using DoE and other analytical data, forming the foundation for method control. [15]
5. Multivariate Analysis and Chemometric Tools:
Analytical data can be complex, especially in spectroscopic or chromatographic techniques. Chemometric and multivariate tools extract meaningful insights, understand variability, and optimize method performance. Examples include:
Applications include method optimization, peak resolution, raw material classification, and robustness evaluation. [15]
6. Control Strategy Tools:
A strong control strategy ensures consistent method performance during routine use. Tools include:
These tools detect deviations early, enabling timely corrective actions. [15]
7. Method Validation Tools:
AQbD-driven validation leverages knowledge gained during development to confirm that the method meets ATP under normal operating conditions. Validation may include:
By integrating method understanding and risk assessment upfront, validation becomes more efficient and scientifically justified. [15]
8. Lifecycle Management Tools:
Lifecycle management ensures methods remain fit-for-purpose throughout their operational life. Tools include:
These tools enable analytical methods to adjust to changing conditions while still ensuring consistent quality and regulatory compliance. [15]
9. Regulatory Frameworks:
AQbD tools are applied in accordance with regulatory expectations. Key frameworks include:
Regulatory tools support scientific justification of method parameters, MODR, and control strategies, enabling regulatory flexibility and acceptance. [4][6]
Analytical Quality through Design: Case Studies
Analytical Quality through Design offers a structured, science driven, and risk based approach for developing analytical methods that are robust, dependable, and consistently meet pre-established performance criteria. In recent years, its use has expanded across pharmaceutical research and quality control settings to build methods that are efficient, reproducible, and aligned with regulatory expectations. The case studies below illustrate real-world applications of AQbD across various analytical techniques, demonstrating how elements such as risk assessment, Design of Experiments, establishment of a Method Operable Design Region, and ongoing lifecycle management enhance method performance and overall reliability. [7][8][9]
Case Study 1: HPLC Technique for Simultaneous Quantification of Drugs
Objective: To establish a stability-indicating HPLC technique that can concurrently quantify multiple active pharmaceutical ingredients (APIs) in a combination formulation, while efficiently resolving them from excipients and potential degradation products.
AQbD Implementation:
Case Study 2: RP-HPLC Method for Forced Degradation Analysis
Objective: To develop a reversed-phase HPLC method for detecting and measuring degradation products formed under various stress conditions, such as acidic, basic, oxidative, thermal, and photolytic stress.
AQbD Implementation:
Case Study 3: LC-MS/MS Technique for Plasma Drug Analysis
Objective: To develop an LC-MS/MS-based bioanalytical approach for precise measurement of drug concentrations in plasma, facilitating pharmacokinetic assessments and bioequivalence studies.
AQbD Implementation:
Case Study 4: UV-Visible Spectrophotometric Method
Objective: To develop a UV-Visible spectrophotometric technique for precise quantification of a single active pharmaceutical ingredient (API) in tablet formulations.
AQbD Implementation:
Case Study 5: Capillary Electrophoresis Method
Objective: To create a capillary electrophoresis method capable of simultaneously measuring multiple active pharmaceutical ingredients (APIs) within a combined dosage form.
AQbD Implementation:
Key Insights from AQbD Case Studies:
CONCLUSION:
Analytical Quality through Design offers a scientific, structured, and risk-based framework for creating analytical methods that are both robust and reliable. The approach begins with establishing the Analytical Target Profile, followed by systematic risk evaluation and the use of tools such as DoE, defining the MODR, and implementing continuous controls strategies. Evidence from various case studies indicates that AQbD based methods provide improved accuracy, precision, and robustness compared to traditional method development. In summary, AQbD strengthens method understanding, minimizes variability, and supports effective lifecycle management, ensuring consistent and high quality results. [3]
REFERENCES
Rasika Tilekar, Smita Waghmare, Neelam Bhagdewani, Prem Jadhav, Bhavana Galande, Pramod Ingale, Analytical Quality by Design (AQbD): Principles, Tools and Case Studies, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 12, 2923-2934. https://doi.org/10.5281/zenodo.17985685
10.5281/zenodo.17985685