<img height="1" width="1" style="display:none;" alt="" src="https://px.ads.linkedin.com/collect/?pid=489233&amp;fmt=gif">

Critical challenges to implementing the QbD methodology

Published
Updated

Quality professionals in the life sciences industry are dedicated to achieving the highest standards of quality, safety, and efficacy in all products, while also improving manufacturability. 

Medicines must meet the intended use. To accomplish this, they employ an effective and systematic approach known as Quality By Design (QbD) methodology that facilitates:

  • Product development
  • Process understanding
  • Process capability
  • Process variability control 
  • Using novel approaches
  • Improving operational performance

By integrating prior knowledge, quality planning, design of experiments results, and risk assessment throughout the product life cycle, organizations ensure new products and services are delivered to satisfy customers' requirements consistently and efficiently.

This methodical framework enables proactive identification and mitigation of potential risks, ultimately leading to optimized processes and consistent, reliable outcomes. 

It is unquestionable that the QbD approach fosters a culture of excellence and compliance. However, despite the benefits though, its implementation is not so easy.

Curious to know the reasons? Keep reading as we delve into the key challenges of implementing QbD, providing you with valuable insights and clues.



 

Introduction to QbD

The concept of QbD was first developed by Dr. Joseph M. Juran, the Father of Quality. He was also the driver behind the use of the Pareto Principle -rule 80/20- and the Quality Trilogy, laying the foundations of Six Sigma and DMAIC. 

QbD represents a systematic and purposeful approach to development, starting with well-defined objectives. It places significant emphasis on gaining in-depth understanding of both product and process, while ensuring meticulous control measures through the lens of scientific rationale and effective risk management. 

While QbD has long been used in various manufacturing sectors, it has gained significant traction in the pharmaceutical industry through the FDA's initiative:  Pharmaceutical CGMP Initiative for the 21st Century – a Risk Based Approach, and through the implementation of ICH guidelines ICH Q8 (Pharmaceutical Development), ICH Q9 (Quality Risk Management), and ICH Q10 (Pharmaceutical Quality Systems).

The success of QbD relies on two critical factors: pharmaceutical development studies and manufacturing experience. These elements provide valuable scientific knowledge, enabling the establishment of the design space, specifications, and manufacturing controls. 

The focus of QbD is that quality cannot be tested into a product. QbD goes beyond the traditional Quality By Testing (QbT), as quality must be built into a product.

QbD is an inherent part of the design. Therefore, any changes in formulation or manufacturing processes should be seen as opportunities to enhance knowledge of the risks involved and reinforce the design space to mitigate those risks.

The design space sets the boundaries of process understanding for normal operations, easing a more flexible regulatory approach. Deviations from the design space trigger a regulatory post-approval process, highlighting the importance of adhering to the established parameters.

The identification and justification of critical quality attributes (CQAs) are based on their significant impact on product quality. By thoroughly understanding these crucial elements, quality managers can implement effective control strategies that ensure consistent product quality.

 

Benefits of QbD

Implementing QbD methodology offers numerous benefits. These include:

  • Streamlining manufacturing process improvements within the approved design space
  • Implementing real-time quality control measures. 
  • Reducing post-approval submissions
  • Making risk-based regulatory decisions

To capitalize on these advantages, quality managers must demonstrate an enhanced knowledge of product performance through techniques like formal Design of Experiments (DoE), process analytical technology (PAT), and the application of quality risk management (QRM) principles.



Key challenges to implement QbD

 

1. Defining QTPPs, CQAs, CPPs, and Control Strategy: 

Establishing the Quality Target Product Profile (QTPP) is challenging due to its complexity and requires careful consideration of quality, safety, and efficacy aspects, such as the route of administration, dosage form, bioavailability, strength, and stability. 

Identifying the Critical Quality Attribute’s (CQAs), and the Critical Process Parameters (CPPs) that obtains drug products are also essential to understand the characteristics that significantly impact its quality consistently. These elements must be monitored and controlled to reduce variability, which can be challenging due to the multitude of factors and potential interactions involved.

 

2. Controlling variability during manufacturing

Designing and controlling manufacturing processes with controlled variability to consistently deliver products with the desired quality attributes is a critical challenge.

Variability can arise from various sources, including quality of raw materials, equipment functioning, environmental conditions, and human factors. 

By mapping out processes and the relationships with QTPPs, CQAs, and CPPs, organizations can determine the critical points in the manufacturing process that need to be tightly controlled to ensure consistent product quality.

Another challenge is the development and implementation of appropriate process monitoring and control systems. It involves the use of advanced technologies and analytical methods to continuously monitor and analyze critical process parameters, allowing for real-time adjustments and interventions when unexpected trends or deviations occur. These systems can include in-line sensors, automated control systems, statistical process control techniques, and other quality assurance tools.

Additionally, controlling variability requires a robust quality management system and strong emphasis on risk assessment and mitigation. By identifying potential sources of variability and implementing proactive measures to mitigate risks, organizations can enhance process control and minimize the likelihood of quality-related issues.

Overall, controlling variability requires a multidisciplinary approach, collaboration between different stakeholders, advanced technologies, and a thorough understanding of the process and its critical parameters. By effectively managing variability, organizations can achieve consistent product quality, improve efficiency, and ensure the safety and efficacy of their products.

 

3. Defining a control strategy

Developing a robust control strategy is essential to ensure the defined quality requirements are consistently met throughout the product lifecycle. 

Professionals who define the strategy require comprehensive knowledge of the CQAs and their relationship to product performance and safety.

This includes determining appropriate control points, monitoring techniques, and acceptance criteria.

 

4. Mindset Shift

Adopting a QbD mindset requires a shift in the traditional approach to pharmaceutical development. 

It involves moving from a reactive mindset focused on quality control testing to a proactive approach centered around designing quality built into the product from the beginning. This shift requires significant quality culture change in organizations.

 

5. Resource Allocation

Implementing QbD requires substantial investment in terms of time and resources.

Adequate resources must be allocated for conducting thorough scientific studies, establishing a robust design space, and implementing advanced analytical techniques. 

This can be a challenge for organizations without the appropriate resources and tools.

 

6. Knowledge and Expertise

QbD implementation demands a high level of scientific knowledge and expertise. It requires a deep understanding of product attributes, process parameters, and the interrelationships between them. 

Acquiring and retaining the necessary expertise within the organization can be a challenge, especially in rapidly evolving scientific and technological landscapes.

 

7. Regulatory Compliance

While regulatory agencies encourage the adoption of QbD, navigating the regulatory landscape can still be challenging. Ensuring compliance with evolving regulatory requirements and effectively communicating the QbD approach to regulatory authorities can pose hurdles.

Additionally, regulatory agencies' expectations regarding the documentation and justification of the design space and control strategies may require careful planning and execution.

 

8. Data Availability and Quality

QbD relies on data-driven decision-making and robust scientific understanding. 

However, obtaining reliable and comprehensive data can be challenging without the proper tools, especially during early stages of development. Ensuring data integrity, accuracy, and consistency is crucial for successful QbD implementation.



Conclusion

While implementing the QbD methodology may present critical challenges, organizations that successfully navigate these hurdles stand to gain substantial advantages in terms of product quality, regulatory compliance, and operational excellence. Embracing the QbD approach will undoubtedly contribute to the continued improvement and advancement of the pharmaceutical industry, ultimately benefiting patients and healthcare providers alike.