Control Strategies

In a past post discussing the program level in the document hierarchy, I outlined how program documents serve as critical connective tissue between high-level policies and detailed procedures. Today, I’ll explore three distinct but related approaches to control strategies: the Annex 1 Contamination Control Strategy (CCS), the ICH Q8 Process Control Strategy, and a Technology Platform Control Strategy. Understanding their differences and relationships allows us to establish a comprehensive quality system in pharmaceutical manufacturing, especially as regulatory requirements continue to evolve and emphasize more scientific, risk-based approaches to quality management.

Control strategies have evolved significantly and are increasingly central to pharmaceutical quality management. As I noted in my previous article, program documents create an essential mapping between requirements and execution, demonstrating the design thinking that underpins our quality processes. Control strategies exemplify this concept, providing comprehensive frameworks that ensure consistent product quality through scientific understanding and risk management.

The pharmaceutical industry has gradually shifted from reactive quality testing to proactive quality design. This evolution mirrors the maturation of our document hierarchies, with control strategies occupying that critical program-level space between overarching quality policies and detailed operational procedures. They serve as the blueprint for how quality will be achieved, maintained, and improved throughout a product’s lifecycle.

This evolution has been accelerated by increasing regulatory scrutiny, particularly following numerous drug recalls and contamination events resulting in significant financial losses for pharmaceutical companies.

Annex 1 Contamination Control Strategy: A Facility-Focused Approach

The Annex 1 Contamination Control Strategy represents a comprehensive, facility-focused approach to preventing chemical, physical and microbial contamination in pharmaceutical manufacturing environments. The CCS takes a holistic view of the entire manufacturing facility rather than focusing on individual products or processes.

A properly implemented CCS requires a dedicated cross-functional team representing technical knowledge from production, engineering, maintenance, quality control, microbiology, and quality assurance. This team must systematically identify contamination risks throughout the facility, develop mitigating controls, and establish monitoring systems that provide early detection of potential issues. The CCS must be scientifically formulated and tailored specifically for each manufacturing facility’s unique characteristics and risks.

What distinguishes the Annex 1 CCS is its infrastructural approach to Quality Risk Management. Rather than focusing solely on product attributes or process parameters, it examines how facility design, environmental controls, personnel practices, material flow, and equipment operate collectively to prevent contamination. The CCS process involves continual identification, scientific evaluation, and effective control of potential contamination risks to product quality.

Critical Factors in Developing an Annex 1 CCS

The development of an effective CCS involves several critical considerations. According to industry experts, these include identifying the specific types of contaminants that pose a risk, implementing appropriate detection methods, and comprehensively understanding the potential sources of contamination. Additionally, evaluating the risk of contamination and developing effective strategies to control and minimize such risks are indispensable components of an efficient contamination control system.

When implementing a CCS, facilities should first determine their critical control points. Annex 1 highlights the importance of considering both plant design and processes when developing a CCS. The strategy should incorporate a monitoring and ongoing review system to identify potential lapses in the aseptic environment and contamination points in the facility. This continuous assessment approach ensures that contamination risks are promptly identified and addressed before they impact product quality.

ICH Q8 Process Control Strategy: The Quality by Design Paradigm

While the Annex 1 CCS focuses on facility-wide contamination prevention, the ICH Q8 Process Control Strategy takes a product-centric approach rooted in Quality by Design (QbD) principles. The ICH Q8(R2) guideline introduces control strategy as “a planned set of controls derived from current product and process understanding that ensures process performance and product quality”. This approach emphasizes designing quality into products rather than relying on final testing to detect issues.

The ICH Q8 guideline outlines a set of key principles that form the foundation of an effective process control strategy. At its core is pharmaceutical development, which involves a comprehensive understanding of the product and its manufacturing process, along with identifying critical quality attributes (CQAs) that impact product safety and efficacy. Risk assessment plays a crucial role in prioritizing efforts and resources to address potential issues that could affect product quality.

The development of an ICH Q8 control strategy follows a systematic sequence: defining the Quality Target Product Profile (QTPP), identifying Critical Quality Attributes (CQAs), determining Critical Process Parameters (CPPs) and Critical Material Attributes (CMAs), and establishing appropriate control methods. This scientific framework enables manufacturers to understand how material attributes and process parameters affect product quality, allowing for more informed decision-making and process optimization.

Design Space and Lifecycle Approach

A unique aspect of the ICH Q8 control strategy is the concept of “design space,” which represents a range of process parameters within which the product will consistently meet desired quality attributes. Developing and demonstrating a design space provides flexibility in manufacturing without compromising product quality. This approach allows manufacturers to make adjustments within the established parameters without triggering regulatory review, thus enabling continuous improvement while maintaining compliance.

What makes the ICH Q8 control strategy distinct is its dynamic, lifecycle-oriented nature. The guideline encourages a lifecycle approach to product development and manufacturing, where continuous improvement and monitoring are carried out throughout the product’s lifecycle, from development to post-approval. This approach creates a feedback-feedforward “controls hub” that integrates risk management, knowledge management, and continuous improvement throughout the product lifecycle.

Technology Platform Control Strategies: Leveraging Prior Knowledge

As pharmaceutical development becomes increasingly complex, particularly in emerging fields like cell and gene therapies, technology platform control strategies offer an approach that leverages prior knowledge and standardized processes to accelerate development while maintaining quality standards. Unlike product-specific control strategies, platform strategies establish common processes, parameters, and controls that can be applied across multiple products sharing similar characteristics or manufacturing approaches.

The importance of maintaining state-of-the-art technology platforms has been highlighted in recent regulatory actions. A January 2025 FDA Warning Letter to Sanofi, concerning a facility that had previously won the ISPE’s Facility of the Year award in 2020, emphasized the requirement for “timely technological upgrades to equipment/facility infrastructure”. This regulatory focus underscores that even relatively new facilities must continually evolve their technological capabilities to maintain compliance and product quality.

Developing a Comprehensive Technology Platform Roadmap

A robust technology platform control strategy requires a well-structured technology roadmap that anticipates both regulatory expectations and technological advancements. According to recent industry guidance, this roadmap should include several key components:

At its foundation, regular assessment protocols are essential. Organizations should conduct comprehensive annual evaluations of platform technologies, examining equipment performance metrics, deviations associated with the platform, and emerging industry standards that might necessitate upgrades. These assessments should be integrated with Facility and Utility Systems Effectiveness (FUSE) metrics and evaluated through structured quality governance processes.

The technology roadmap must also incorporate systematic methods for monitoring industry trends. This external vigilance ensures platform technologies remain current with evolving expectations and capabilities.

Risk-based prioritization forms another critical element of the platform roadmap. By utilizing living risk assessments, organizations can identify emerging issues and prioritize platform upgrades based on their potential impact on product quality and patient safety. These assessments should represent the evolution of the original risk management that established the platform, creating a continuous thread of risk evaluation throughout the platform’s lifecycle.

Implementation and Verification of Platform Technologies

Successful implementation of platform technologies requires robust change management procedures. These should include detailed documentation of proposed platform modifications, impact assessments on product quality across the portfolio, appropriate verification activities, and comprehensive training programs. This structured approach ensures that platform changes are implemented systematically with full consideration of their potential implications.

Verification activities for platform technologies must be particularly thorough, given their application across multiple products. The commissioning, qualification, and validation activities should demonstrate not only that platform components meet predetermined specifications but also that they maintain their intended performance across the range of products they support. This verification must consider the variability in product-specific requirements while confirming the platform’s core capabilities.

Continuous monitoring represents the final essential element of platform control strategies. By implementing ongoing verification protocols aligned with Stage 3 of the FDA’s process validation model, organizations can ensure that platform technologies remain in a state of control during routine commercial manufacture. This monitoring should anticipate and prevent issues, detect unplanned deviations, and identify opportunities for platform optimization.

Leveraging Advanced Technologies in Platform Strategies

Modern technology platforms increasingly incorporate advanced capabilities that enhance their flexibility and performance. Single-Use Systems (SUS) reduce cleaning and validation requirements while improving platform adaptability across products. Modern Microbial Methods (MMM) offer advantages over traditional culture-based approaches in monitoring platform performance. Process Analytical Technology (PAT) enables real-time monitoring and control, enhancing product quality and process understanding across the platform. Data analytics and artificial intelligence tools identify trends, predict maintenance needs, and optimize processes across the product portfolio.

The implementation of these advanced technologies within platform strategies creates significant opportunities for standardization, knowledge transfer, and continuous improvement. By establishing common technological foundations that can be applied across multiple products, organizations can accelerate development timelines, reduce validation burdens, and focus resources on understanding the unique aspects of each product while maintaining a robust quality foundation.

How Control Strategies Tie Together Design, Qualification/Validation, and Risk Management

Control strategies serve as the central nexus connecting design, qualification/validation, and risk management in a comprehensive quality framework. This integration is not merely beneficial but essential for ensuring product quality while optimizing resources. A well-structured control strategy creates a coherent narrative from initial concept through on-going production, ensuring that design intentions are preserved through qualification activities and ongoing risk management.

During the design phase, scientific understanding of product and process informs the development of the control strategy. This strategy then guides what must be qualified and validated and to what extent. Rather than validating everything (which adds cost without necessarily improving quality), the control strategy directs validation resources toward aspects most critical to product quality.

The relationship works in both directions—design decisions influence what will require validation, while validation capabilities and constraints may inform design choices. For example, a process designed with robust, well-understood parameters may require less extensive validation than one operating at the edge of its performance envelope. The control strategy documents this relationship, providing scientific justification for validation decisions based on product and process understanding.

Risk management principles are foundational to modern control strategies, informing both design decisions and priorities. A systematic risk assessment approach helps identify which aspects of a process or facility pose the greatest potential impact on product quality and patient safety. The control strategy then incorporates appropriate controls and monitoring systems for these high-risk elements, ensuring that validation efforts are proportionate to risk levels.

The Feedback-Feedforward Mechanism

One of the most powerful aspects of an integrated control strategy is its ability to function as what experts call a feedback-feedforward controls hub. As a product moves through its lifecycle, from development to commercial manufacturing, the control strategy evolves based on accumulated knowledge and experience. Validation results, process monitoring data, and emerging risks all feed back into the control strategy, which in turn drives adjustments to design parameters and validation approaches.

Comparing Control Strategy Approaches: Similarities and Distinctions

While these three control strategy approaches have distinct focuses and applications, they share important commonalities. All three emphasize scientific understanding, risk management, and continuous improvement. They all serve as program-level documents that connect high-level requirements with operational execution. And all three have gained increasing regulatory recognition as pharmaceutical quality management has evolved toward more systematic, science-based approaches.

AspectAnnex 1 CCSICH Q8 Process Control StrategyTechnology Platform Control Strategy
Primary FocusFacility-wide contamination preventionProduct and process qualityStandardized approach across multiple products
ScopeMicrobial, pyrogen, and particulate contamination (a good one will focus on physical, chemical and biologic hazards)All aspects of product qualityCommon technology elements shared across products
Regulatory FoundationEU GMP Annex 1 (2022 revision)ICH Q8(R2)Emerging FDA guidance (Platform Technology Designation)
Implementation LevelManufacturing facilityIndividual productTechnology group or platform
Key ComponentsContamination risk identification, detection methods, understanding of contamination sourcesQTPP, CQAs, CPPs, CMAs, design spaceStandardized technologies, processes, and controls
Risk Management ApproachInfrastructural (facility design, processes, personnel) – great for a HACCPProduct-specific (process parameters, material attributes)Platform-specific (shared technological elements)
Team StructureCross-functional (production, engineering, QC, QA, microbiology)Product development, manufacturing and qualityTechnology development and product adaptation
Lifecycle ConsiderationsContinuous monitoring and improvement of facility controlsProduct lifecycle from development to post-approvalEvolution of platform technology across multiple products
DocumentationFacility-specific CCS with ongoing monitoring recordsProduct-specific control strategy with design space definitionPlatform master file with product-specific adaptations
FlexibilityLow (facility-specific controls)Medium (within established design space)High (adaptable across multiple products)
Primary BenefitContamination prevention and controlConsistent product quality through scientific understandingEfficiency and knowledge leverage across product portfolio
Digital IntegrationEnvironmental monitoring systems, facility controlsProcess analytical technology, real-time release testingPlatform data management and cross-product analytics

These approaches are not mutually exclusive; rather, they complement each other within a comprehensive quality management system. A manufacturing site producing sterile products needs both an Annex 1 CCS for facility-wide contamination control and ICH Q8 process control strategies for each product. If the site uses common technology platforms across multiple products, platform control strategies would provide additional efficiency and standardization.

Control Strategies Through the Lens of Knowledge Management: Enhancing Quality and Operational Excellence

The pharmaceutical industry’s approach to control strategies has evolved significantly in recent years, with systematic knowledge management emerging as a critical foundation for their effectiveness. Control strategies—whether focused on contamination prevention, process control, or platform technologies—fundamentally depend on how knowledge is created, captured, disseminated, and applied across an organization. Understanding the intersection between control strategies and knowledge management provides powerful insights into building more robust pharmaceutical quality systems and achieving higher levels of operational excellence.

The Knowledge Foundation of Modern Control Strategies

Control strategies represent systematic approaches to ensuring consistent pharmaceutical quality by managing various aspects of production. While these strategies differ in focus and application, they share a common foundation in knowledge—both explicit (documented) and tacit (experiential).

Knowledge Management as the Binding Element

The ICH Q10 Pharmaceutical Quality System model positions knowledge management alongside quality risk management as dual enablers of pharmaceutical quality. This pairing is particularly significant when considering control strategies, as it establishes what might be called a “Risk-Knowledge Infinity Cycle”—a continuous process where increased knowledge leads to decreased uncertainty and therefore decreased risk. Control strategies represent the formal mechanisms through which this cycle is operationalized in pharmaceutical manufacturing.

Effective control strategies require comprehensive knowledge visibility across functional areas and lifecycle phases. Organizations that fail to manage knowledge effectively often experience problems like knowledge silos, repeated issues due to lessons not learned, and difficulty accessing expertise or historical product knowledge—all of which directly impact the effectiveness of control strategies and ultimately product quality.

The Feedback-Feedforward Controls Hub: A Knowledge Integration Framework

As described above, the heart of effective control strategies lies is the “feedback-feedforward controls hub.” This concept represents the integration point where knowledge flows bidirectionally to continuously refine and improve control mechanisms. In this model, control strategies function not as static documents but as dynamic knowledge systems that evolve through continuous learning and application.

The feedback component captures real-time process data, deviations, and outcomes that generate new knowledge about product and process performance. The feedforward component takes this accumulated knowledge and applies it proactively to prevent issues before they occur. This integrated approach creates a self-reinforcing cycle where control strategies become increasingly sophisticated and effective over time.

For example, in an ICH Q8 process control strategy, process monitoring data feeds back into the system, generating new understanding about process variability and performance. This knowledge then feeds forward to inform adjustments to control parameters, risk assessments, and even design space modifications. The hub serves as the central coordination mechanism ensuring these knowledge flows are systematically captured and applied.

Knowledge Flow Within Control Strategy Implementation

Knowledge flows within control strategies typically follow the knowledge management process model described in the ISPE Guide, encompassing knowledge creation, curation, dissemination, and application. For control strategies to function effectively, this flow must be seamless and well-governed.

The systematic management of knowledge within control strategies requires:

  1. Methodical capture of knowledge through various means appropriate to the control strategy context
  2. Proper identification, review, and analysis of this knowledge to generate insights
  3. Effective storage and visibility to ensure accessibility across the organization
  4. Clear pathways for knowledge application, transfer, and growth

When these elements are properly integrated, control strategies benefit from continuous knowledge enrichment, resulting in more refined and effective controls. Conversely, barriers to knowledge flow—such as departmental silos, system incompatibilities, or cultural resistance to knowledge sharing—directly undermine the effectiveness of control strategies.

Annex 1 Contamination Control Strategy Through a Knowledge Management Lens

The Annex 1 Contamination Control Strategy represents a facility-focused approach to preventing microbial, pyrogen, and particulate contamination. When viewed through a knowledge management lens, the CCS becomes more than a compliance document—it emerges as a comprehensive knowledge system integrating multiple knowledge domains.

Effective implementation of an Annex 1 CCS requires managing diverse knowledge types across functional boundaries. This includes explicit knowledge documented in environmental monitoring data, facility design specifications, and cleaning validation reports. Equally important is tacit knowledge held by personnel about contamination risks, interventions, and facility-specific nuances that are rarely fully documented.

The knowledge management challenges specific to contamination control include ensuring comprehensive capture of contamination events, facilitating cross-functional knowledge sharing about contamination risks, and enabling access to historical contamination data and prior knowledge. Organizations that approach CCS development with strong knowledge management practices can create living documents that continuously evolve based on accumulated knowledge rather than static compliance tools.

Knowledge mapping is particularly valuable for CCS implementation, helping to identify critical contamination knowledge sources and potential knowledge gaps. Communities of practice spanning quality, manufacturing, and engineering functions can foster collaboration and tacit knowledge sharing about contamination control. Lessons learned processes ensure that insights from contamination events contribute to continuous improvement of the control strategy.

ICH Q8 Process Control Strategy: Quality by Design and Knowledge Management

The ICH Q8 Process Control Strategy embodies the Quality by Design paradigm, where product and process understanding drives the development of controls that ensure consistent quality. This approach is fundamentally knowledge-driven, making effective knowledge management essential to its success.

The QbD approach begins with applying prior knowledge to establish the Quality Target Product Profile (QTPP) and identify Critical Quality Attributes (CQAs). Experimental studies then generate new knowledge about how material attributes and process parameters affect these quality attributes, leading to the definition of a design space and control strategy. This sequence represents a classic knowledge creation and application cycle that must be systematically managed.

Knowledge management challenges specific to ICH Q8 process control strategies include capturing the scientific rationale behind design choices, maintaining the connectivity between risk assessments and control parameters, and ensuring knowledge flows across development and manufacturing boundaries. Organizations that excel at knowledge management can implement more robust process control strategies by ensuring comprehensive knowledge visibility and application.

Particularly important for process control strategies is the management of decision rationale—the often-tacit knowledge explaining why certain parameters were selected or why specific control approaches were chosen. Explicit documentation of this decision rationale ensures that future changes to the process can be evaluated with full understanding of the original design intent, avoiding unintended consequences.

Technology Platform Control Strategies: Leveraging Knowledge Across Products

Technology platform control strategies represent standardized approaches applied across multiple products sharing similar characteristics or manufacturing technologies. From a knowledge management perspective, these strategies exemplify the power of knowledge reuse and transfer across product boundaries.

The fundamental premise of platform approaches is that knowledge gained from one product can inform the development and control of similar products, creating efficiencies and reducing risks. This depends on robust knowledge management practices that make platform knowledge visible and available across product teams and lifecycle phases.

Knowledge management challenges specific to platform control strategies include ensuring consistent knowledge capture across products, facilitating cross-product learning, and balancing standardization with product-specific requirements. Organizations with mature knowledge management practices can implement more effective platform strategies by creating knowledge repositories, communities of practice, and lessons learned processes that span product boundaries.

Integrating Control Strategies with Design, Qualification/Validation, and Risk Management

Control strategies serve as the central nexus connecting design, qualification/validation, and risk management in a comprehensive quality framework. This integration is not merely beneficial but essential for ensuring product quality while optimizing resources. A well-structured control strategy creates a coherent narrative from initial concept through commercial production, ensuring that design intentions are preserved through qualification activities and ongoing risk management.

The Design-Validation Continuum

Control strategies form a critical bridge between product/process design and validation activities. During the design phase, scientific understanding of the product and process informs the development of the control strategy. This strategy then guides what must be validated and to what extent. Rather than validating everything (which adds cost without necessarily improving quality), the control strategy directs validation resources toward aspects most critical to product quality.

The relationship works in both directions—design decisions influence what will require validation, while validation capabilities and constraints may inform design choices. For example, a process designed with robust, well-understood parameters may require less extensive validation than one operating at the edge of its performance envelope. The control strategy documents this relationship, providing scientific justification for validation decisions based on product and process understanding.

Risk-Based Prioritization

Risk management principles are foundational to modern control strategies, informing both design decisions and validation priorities. A systematic risk assessment approach helps identify which aspects of a process or facility pose the greatest potential impact on product quality and patient safety. The control strategy then incorporates appropriate controls and monitoring systems for these high-risk elements, ensuring that validation efforts are proportionate to risk levels.

The Feedback-Feedforward Mechanism

The feedback-feedforward controls hub represents a sophisticated integration of two fundamental control approaches, creating a central mechanism that leverages both reactive and proactive control strategies to optimize process performance. This concept emerges as a crucial element in modern control systems, particularly in pharmaceutical manufacturing, chemical processing, and advanced mechanical systems.

To fully grasp the concept of a feedback-feedforward controls hub, we must first distinguish between its two primary components. Feedback control works on the principle of information from the outlet of a process being “fed back” to the input for corrective action. This creates a loop structure where the system reacts to deviations after they occur. Fundamentally reactive in nature, feedback control takes action only after detecting a deviation between the process variable and setpoint.

In contrast, feedforward control operates on the principle of preemptive action. It monitors load variables (disturbances) that affect a process and takes corrective action before these disturbances can impact the process variable. Rather than waiting for errors to manifest, feedforward control uses data from load sensors to predict when an upset is about to occur, then feeds that information forward to the final control element to counteract the load change proactively.

The feedback-feedforward controls hub serves as a central coordination point where these two control strategies converge and complement each other. As a product moves through its lifecycle, from development to commercial manufacturing, this control hub evolves based on accumulated knowledge and experience. Validation results, process monitoring data, and emerging risks all feed back into the control strategy, which in turn drives adjustments to design parameters and validation approaches.

Knowledge Management Maturity in Control Strategy Implementation

The effectiveness of control strategies is directly linked to an organization’s knowledge management maturity. Organizations with higher knowledge management maturity typically implement more robust, science-based control strategies that evolve effectively over time. Conversely, organizations with lower maturity often struggle with static control strategies that fail to incorporate learning and experience.

Common knowledge management gaps affecting control strategies include:

  1. Inadequate mechanisms for capturing tacit knowledge from subject matter experts
  2. Poor visibility of knowledge across organizational and lifecycle boundaries
  3. Ineffective lessons learned processes that fail to incorporate insights into control strategies
  4. Limited knowledge sharing between sites implementing similar control strategies
  5. Difficulty accessing historical knowledge that informed original control strategy design

Addressing these gaps through systematic knowledge management practices can significantly enhance control strategy effectiveness, leading to more robust processes, fewer deviations, and more efficient responses to change.

The examination of control strategies through a knowledge management lens reveals their fundamentally knowledge-dependent nature. Whether focused on contamination control, process parameters, or platform technologies, control strategies represent the formal mechanisms through which organizational knowledge is applied to ensure consistent pharmaceutical quality.

Organizations seeking to enhance their control strategy effectiveness should consider several key knowledge management principles:

  1. Recognize both explicit and tacit knowledge as essential components of effective control strategies
  2. Ensure knowledge flows seamlessly across functional boundaries and lifecycle phases
  3. Address all four pillars of knowledge management—people, process, technology, and governance
  4. Implement systematic methods for capturing lessons and insights that can enhance control strategies
  5. Foster a knowledge-sharing culture that supports continuous learning and improvement

By integrating these principles into control strategy development and implementation, organizations can create more robust, science-based approaches that continuously evolve based on accumulated knowledge and experience. This not only enhances regulatory compliance but also improves operational efficiency and product quality, ultimately benefiting patients through more consistent, high-quality pharmaceutical products.

The feedback-feedforward controls hub concept represents a particularly powerful framework for thinking about control strategies, emphasizing the dynamic, knowledge-driven nature of effective controls. By systematically capturing insights from process performance and proactively applying this knowledge to prevent issues, organizations can create truly learning control systems that become increasingly effective over time.

Conclusion: The Central Role of Control Strategies in Pharmaceutical Quality Management

Control strategies—whether focused on contamination prevention, process control, or technology platforms—serve as the intellectual foundation connecting high-level quality policies with detailed operational procedures. They embody scientific understanding, risk management decisions, and continuous improvement mechanisms in a coherent framework that ensures consistent product quality.

Regulatory Needs and Control Strategies

Regulatory guidelines like ICH Q8 and Annex 1 CCS underscore the importance of control strategies in ensuring product quality and compliance. ICH Q8 emphasizes a Quality by Design (QbD) approach, where product and process understanding drives the development of controls. Annex 1 CCS focuses on facility-wide contamination prevention, highlighting the need for comprehensive risk management and control systems. These regulatory expectations necessitate robust control strategies that integrate scientific knowledge with operational practices.

Knowledge Management: The Backbone of Effective Control Strategies

Knowledge management (KM) plays a pivotal role in the effectiveness of control strategies. By systematically acquiring, analyzing, storing, and disseminating information related to products and processes, organizations can ensure that the right knowledge is available at the right time. This enables informed decision-making, reduces uncertainty, and ultimately decreases risk.

Risk Management and Control Strategies

Risk management is inextricably linked with control strategies. By identifying and mitigating risks, organizations can maintain a state of control and facilitate continual improvement. Control strategies must be designed to incorporate risk assessments and management processes, ensuring that they are proactive and adaptive.

The Interconnectedness of Control Strategies

Control strategies are not isolated entities but are interconnected with design, qualification/validation, and risk management processes. They form a feedback-feedforward controls hub that evolves over a product’s lifecycle, incorporating new insights and adjustments based on accumulated knowledge and experience. This dynamic approach ensures that control strategies remain effective and relevant, supporting both regulatory compliance and operational excellence.

Why Control Strategies Are Key

Control strategies are essential for several reasons:

  1. Regulatory Compliance: They ensure adherence to regulatory guidelines and standards, such as ICH Q8 and Annex 1 CCS.
  2. Quality Assurance: By integrating scientific understanding and risk management, control strategies guarantee consistent product quality.
  3. Operational Efficiency: Effective control strategies streamline processes, reduce waste, and enhance productivity.
  4. Knowledge Management: They facilitate the systematic management of knowledge, ensuring that insights are captured and applied across the organization.
  5. Risk Mitigation: Control strategies proactively identify and mitigate risks, protecting both product quality and patient safety.

Control strategies represent the central mechanism through which pharmaceutical companies ensure quality, manage risk, and leverage knowledge. As the industry continues to evolve with new technologies and regulatory expectations, the importance of robust, science-based control strategies will only grow. By integrating knowledge management, risk management, and regulatory compliance, organizations can develop comprehensive quality systems that protect patients, satisfy regulators, and drive operational excellence.

Critical Material Attributes

In the complex landscape of biologics drug substance (DS) manufacturing, the understanding and management of Critical Material Attributes (CMAs) has emerged as a cornerstone for achieving consistent product quality. As biological products represent increasingly sophisticated therapeutic modalities with intricate structural characteristics and manufacturing processes, the identification and control of CMAs become vital components of a robust Quality by Design (QbD) approach. It is important to have a strong process for the selection, risk management, and qualification/validation of CMAs, capturing their relationships with Critical Quality Attributes (CQAs) and Critical Process Parameters (CPPs).

Defining Critical Material Attributes

Critical Material Attributes (CMA) represent a fundamental concept within the pharmaceutical QbD paradigm. A CMA is a physical, chemical, biological, or microbiological property or characteristic of an input material controlled within an appropriate limit, range, or distribution to ensure the desired quality of output material. While not officially codified in guidance, this definition has become widely accepted throughout the industry as an essential concept for implementing QbD principles in biotech manufacturing.

In biologics drug substance manufacturing, CMAs may encompass attributes of raw materials used in cell culture media, chromatography resins employed in purification steps, and various other input materials that interact with the biological product during production. For example, variations in the composition of cell culture media components can significantly impact cell growth kinetics, post-translational modifications, and, ultimately, the critical quality attributes of the final biological product.

The biologics manufacturing process typically encompasses both upstream processing (USP) and downstream processing (DSP) operations. Within this continuum, product development aims to build robustness and demonstrate control of a manufacturing process to ensure consistency within the specifications of the manufacturing quality attributes. QbD principles reinforce the need for a systematic process development approach and risk assessment to be conducted early and throughout the biologics development process.

The Interdependent Relationship: CMAs, CQAs, and CPPs in Biologics Manufacturing

In biologics DS manufacturing, the relationship between CMAs, CPPs, and CQAs forms a complex network that underpins product development and manufacture. CQAs are physical, chemical, biological, or microbiological properties or characteristics of the output product that should remain within appropriate limits to ensure product quality. For biologics, these might include attributes like glycosylation patterns, charge variants, aggregation propensity, or potency—all of which directly impact patient safety and efficacy.

The intricate relationship between these elements in biologics production can or exabe expressed as: CQAs = f(CPP₁, CPP₂, CPP₃, …, CMA₁, CMA₂, CMA₃, …). This formulation crystallizes the understanding that CQAs in a biological product are a function of both process parameters and material attributes. For example, in monoclonal antibody production, glycosylation profiles (a CQA) might be influenced by bioreactor temperature and pH (CPPs) as well as the quality and composition of cell culture media components (CMAs).

Identifying CMAs in manufacturing must be aligned with biopharmaceutical development and manufacturing strategies guided by the product’s Target Product Profile (TPP). QbD principles are applied from the onset of product definition and development to ensure that the product meets patient needs and efficacy requirements. Critical sources of variability are identified and controlled through appropriate control strategies to consistently meet product CQAs, and the process is continually monitored, evaluated, and updated to maintain product quality throughout its life cycle.

The interdependence between unit operations adds another layer of complexity. The output from one unit operation becomes the input for the next, creating a chain of interdependent processes where material attributes at each stage can influence subsequent steps. For example, the transition from upstream cell culture to downstream purification operations where the characteristics of the harvested cell culture fluid significantly impact purification efficiency and product quality.

Systematic Approach to CMA Selection in Biologics Manufacturing

Identifying and selecting CMAs in biologics DS manufacturing represents a methodical process requiring scientific rigor and risk-based decision-making. This process typically begins with establishing a Quality Target Product Profile (QTPP), which outlines the desired quality characteristics of the final biological product, taking into account safety and efficacy considerations.

The first step in CMA selection involves comprehensive material characterization to identify all potentially relevant attributes of input materials used in production. This might include characteristics like purity, solubility, or bioactivity for cell culture media components. For chromatography resins in downstream processing, attributes such as binding capacity, selectivity, or stability might be considered. This extensive characterization creates a foundation of knowledge about the materials that will be used in the biological product’s manufacturing process.

Risk assessment tools play a crucial role in the initial screening of potential CMAs. These might include Failure Mode and Effects Analysis (FMEA), Preliminary Hazards Analysis (PHA), or cause-and-effect matrices that relate material attributes to CQAs.

Once potential high-risk material attributes are identified, experimental studies, often employing the Design of Experiments (DoE) methodology, are conducted to determine whether these attributes genuinely impact CQAs of the biological product and, therefore, should be classified as critical. This empirical verification is essential, as theoretical risk assessments must be confirmed through actual data before final classification as a CMA. The process characterization strategy typically aims to identify process parameters that impact product quality and yield by identifying interactions between process parameters and critical quality attributes, justifying and, if necessary, adjusting manufacturing operating ranges and acceptance criteria, ensuring that the process delivers a product with reproducible yields and purity, and enabling heads-up detection of manufacturing deviations using the established control strategy and knowledge about the impact of process inputs on product quality.

Risk Management Strategies for CMAs in Biologics DS Manufacturing

Risk management for Critical Material Attributes (CMAs) in biologics manufacturing extends far beyond mere identification to encompass a comprehensive strategy for controlling and mitigating risks throughout the product lifecycle. The risk management process typically follows a structured approach comprising risk identification, assessment, control, communication, and review—all essential elements for ensuring biologics quality and safety.

Structured Risk Assessment Methodologies

The first phase in effective CMA risk management involves establishing a cross-functional team to conduct systematic risk assessments. A comprehensive Raw Material Risk Assessment (RMRA) requires input from diverse experts including Manufacturing, Quality Assurance, Quality Control, Supply Chain, and Materials Science & Technology (MSAT) teams, with additional Subject Matter Experts (SMEs) added as necessary. This multidisciplinary approach ensures that diverse perspectives on material criticality are considered, particularly important for complex biologics manufacturing where materials may impact multiple aspects of the process.

Risk assessment methodologies for CMAs must be standardized yet adaptable to different material types. A weight-based scoring system can be implemented where risk criteria are assigned predetermined weights based on the severity that risk realization would pose on the product/process. This approach recognizes that not all material attributes carry equal importance in terms of their potential impact on product quality and patient safety.

Comprehensive Risk Evaluation Categories

When evaluating CMAs, three major categories of risk attributes should be systematically assessed:

  1. User Requirements: These evaluate how the material is used within the manufacturing process and include assessment of:
    • Patient exposure (direct vs. indirect material contact)
    • Impact to product quality (immediate vs. downstream effects)
    • Impact to process performance and consistency
    • Microbial restrictions for the material
    • Regulatory and compendial requirements
    • Material acceptance requirements
  2. Material Attributes: These assess the inherent properties of the material itself:
    • Microbial characteristics and bioburden risk
    • Origin, composition, and structural complexity
    • Material shelf-life and stability characteristics
    • Manufacturing complexity and potential impurities
    • Analytical complexity and compendial status
    • Material handling requirements
  3. Supplier Attributes: These evaluate the supply chain risks associated with the material:
    • Supplier quality system performance
    • Continuity of supply assurance
    • Supplier technical capabilities
    • Supplier relationship and communication
    • Material grade specificity (pharmaceutical vs. industrial)

In biologics manufacturing, these categories take on particular significance. For instance, materials derived from animal sources might carry higher risks related to adventitious agents, while complex cell culture media components might exhibit greater variability in composition between suppliers—both scenarios with potentially significant impacts on product quality.

Quantitative Risk Scoring and Prioritization

Risk assessment for CMAs should employ quantitative scoring methodologies that allow for consistency in evaluation and clear prioritization of risk mitigation activities. For example, risk attributes can be qualitatively scaled as High, Medium, and Low, but then converted to numerical values (High=9, Medium=3, Low=1) to create an adjusted score. These adjusted scores are then multiplied by predetermined weights for each risk criterion to calculate weighted scores.

The total risk score for each raw material is calculated by adding all the weighted scores across categories. This quantitative approach enables objective classification of materials into risk tiers: Low (≤289), Medium (290-600), or High (≥601). Such tiered classification drives appropriate resource allocation, focusing intensified control strategies on truly critical materials while avoiding unnecessary constraints on low-risk items.

This methodology aligns with the QbD principle that not all quality attributes result in the same level of harm to patients, and therefore not all require the same level of control. The EMA-FDA QbD Pilot program emphasized that “the fact that a risk of failure is mitigated by applying a robust proactive control strategy should not allow for the underestimation of assigning criticality.” This suggests that even when control strategies are in place, the fundamental criticality of material attributes should be acknowledged and appropriately managed.

Risk Mitigation Strategies and Control Implementation

For materials identified as having medium to high risk, formalizing mitigation strategies becomes crucial. The level of mitigation required should be proportionate to the risk score. Any material with a Total Risk Score of Medium (290-600) requires a documented mitigation strategy, while materials with High risk scores (≥601) should undergo further evaluation under formal Quality Risk Management procedures. For particularly high-risk materials, consideration should be given to including them on the organization’s risk register to ensure ongoing visibility and management attention.

Mitigation strategies for high-risk CMAs in biologics manufacturing might include:

  1. Enhanced supplier qualification and management programs: For biotech manufacturing, this might involve detailed audits of suppliers’ manufacturing facilities, particularly focusing on areas that could impact critical material attributes such as cell culture media components or chromatography resins.
  2. Tightened material specifications: Implementing more stringent specifications for critical attributes of high-risk materials. For example, for a critical growth factor in cell culture media, the purity, potency, and stability specifications might be tightened beyond the supplier’s standard specifications.
  3. Increased testing frequency: Implementing more frequent or extensive testing protocols for high-risk materials, potentially including lot-to-lot testing for biological activity or critical physical attributes.
  4. Secondary supplier qualification: Developing and qualifying alternative suppliers for high-risk materials to mitigate supply chain disruptions. This is particularly important for specialized biologics materials that may have limited supplier options.
  5. Process modifications to accommodate material variability: Developing processes that can accommodate expected variability in critical material attributes, such as adjustments to cell culture parameters based on growth factor potency measurements.

Continuous Monitoring and Periodic Reassessment

A crucial aspect of CMA risk management in biologics manufacturing is that the risk assessment is not a one-time activity but a continuous process. The RMRA should be treated as a “living document” that requires updating when conditions change or when mitigation efforts reduce the risk associated with a material. At minimum, periodical re-evaluation of the risk assessment should be conducted in accordance with the organization’s Quality Risk Management procedures.

Changes that might trigger reassessment include:

  • Supplier changes or manufacturing site transfers
  • Changes in material composition or manufacturing process
  • New information about material impact on product quality
  • Observed variability in process performance potentially linked to material attributes
  • Regulatory changes affecting material requirements

This continual reassessment approach is particularly important in biologics manufacturing, where understanding of process-product relationships evolves throughout the product lifecycle, and where subtle changes in materials can have magnified effects on biological systems.

The integration of material risk assessments with broader process risk assessments is also essential. The RMRA should be conducted prior to Process Characterization risk assessments to determine whether any raw materials will need to be included in robustness studies. This integration ensures that the impact of material variability on process performance and product quality is systematically evaluated and controlled.

Through this comprehensive approach to risk management for CMAs, biotech manufacturers can develop robust control strategies that ensure consistent product quality while effectively managing the inherent variability and complexity of production systems and their input materials.

Qualification and Validation of CMAs

The qualification and validation of CMAs represent critical steps in translating scientific understanding into practical control strategies for biotech manufacturing. Qualification involves establishing that the analytical methods used to measure CMAs are suitable for their intended purpose, providing accurate and reliable results. This is particularly important for biologics given their complexity and the sophisticated analytical methods required for their characterization.

For biologics DS manufacturing, a comprehensive analytical characterization package is critical for managing process or facility changes in the development cycle. As part of creating the manufacturing process, analytical tests capable of qualitatively and quantitatively characterizing the physicochemical, biophysical, and bioactive/functional potency attributes of the active biological DS are essential. These tests should provide information about the identity (primary and higher order structures), concentration, purity, and in-process impurities (residual host cell protein, mycoplasma, bacterial and adventitious agents, nucleic acids, and other pathogenic viruses).

Validation of CMAs encompasses demonstrating the relationship between these attributes and CQAs through well-designed experiments. This validation process often employs DoE approaches to establish the functional relationship between CMAs and CQAs, quantifying how variations in material attributes influence the final product quality. For example, in a biologics manufacturing context, a DoE study might investigate how variations in the quality of a chromatography resin affect the purity profile of the final drug substance.

Control strategies for validated CMAs might include a combination of raw material specifications, in-process controls, and process parameter adjustments to accommodate material variability. The implementation of control strategies for CMAs should follow a risk-based approach, focusing the most stringent controls on attributes with the highest potential impact on product quality. This prioritization ensures efficient resource allocation while maintaining robust protection against quality failures.

Integrated Control Strategy for CMAs

The culmination of CMA identification, risk assessment, and validation leads to developing an integrated control strategy within the QbD framework for biotech DS manufacturing. This control strategy encompasses the totality of controls implemented to ensure consistent product quality, including specifications for drug substances, raw materials, and controls for each manufacturing process step.

For biologics specifically, robust and optimized analytical assays and characterization methods with well-documented procedures facilitate smooth technology transfer for process development and cGMP manufacturing. A comprehensive analytical characterization package is also critical for managing process or facility changes in the biological development cycle. Such “comparability studies” are key to ensuring that a manufacturing process change will not adversely impact the quality, safety (e.g., immunogenicity), or efficacy of a biologic product.
Advanced monitoring techniques like Process Analytical Technology (PAT) can provide real-time information about material attributes throughout the biologics manufacturing process, enabling immediate corrective actions when variations are detected. This approach aligns with the QbD principle of continual monitoring, evaluation, and updating of the process to maintain product quality throughout its lifecycle.

The typical goal of a Process Characterization Strategy in biologics manufacturing is to identify process parameters that impact product quality and yield by identifying interactions between process parameters and critical quality attributes, justifying and, if necessary, adjusting manufacturing operating ranges and acceptance criteria, ensuring that the process delivers a product with reproducible yields and purity, and enabling early detection of manufacturing deviations using the established control strategy.

Biologics-Specific Considerations in CMA Management

Biologics manufacturing presents unique challenges for CMA management due to biological systems’ inherent complexity and variability. Unlike small molecules, biologics are produced by living cells and undergo complex post-translational modifications that can significantly impact their safety and efficacy. This biological variability necessitates specialized approaches to CMA identification and control.

In biologics DS manufacturing, yield optimization is a significant consideration. Yield refers to downstream efficiency and is the ratio of the mass (weight) of the final purified protein relative to its mass at the start of purification (output/content from upstream bioprocessing). To achieve a high-quality, safe biological product, it is important that the Downstream Processing (DSP) unit operations can efficiently remove all in-process impurities (Host Cell Proteins, nucleic acid, adventitious agents).

The analytical requirements for biologics add another layer of complexity to CMA management. For licensing biopharmaceuticals, development and validation of assays for lot release and stability testing must be included in the specifications for the DS. Most importantly, a potency assay is required that measures the product’s ability to elicit a specific response in a disease-relevant system. This analytical complexity underscores the importance of robust analytical method development for accurately measuring and controlling CMAs.

Conclusion

Critical Material Attributes represent a vital component in the modern pharmaceutical development paradigm. Their systematic identification, risk management, and qualification underpin successful QbD implementation and ensure consistent production of high-quality biological products. By understanding the intricate relationships between CMAs, CPPs, and CQAs, biologics developers can build robust control strategies that accommodate material variability while consistently delivering products that meet their quality targets.

As manufacturing continues to evolve toward more predictive and science-based approaches, the importance of understanding and controlling CMAs will only increase. Future advancements may include improved predictive models linking material attributes to biological product performance, enhanced analytical techniques for real-time monitoring of CMAs, and more sophisticated control strategies that adapt to material variability through automated process adjustments.

The journey from raw to finished products traverses a complex landscape where material attributes interact with process parameters to determine final product quality. By mastering the science of CMAs, developers, and manufacturers can confidently navigate this landscape, ensuring that patients receive safe, effective, and consistent biological medicines. Through continued refinement of these approaches and collaborative efforts between industry and regulatory agencies, biotech manufacturing can further enhance product quality while improving manufacturing efficiency and regulatory compliance.

Sources

APA Bibliography

World Health Organization. (n.d.). Quality risk management (WHO Technical Report Series, No. 981, Annex 2). https://www.who.int/docs/default-source/medicines/norms-and-standards/guidelines/production/trs981-annex2-who-quality-risk-management.pdf

Reducing Subjectivity in Quality Risk Management: Aligning with ICH Q9(R1)

In a previous post, I discussed how overcoming subjectivity in risk management and decision-making requires fostering a culture of quality and excellence. This is an issue that it is important to continue to evaluate and push for additional improvement.

The revised ICH Q9(R1) guideline, finalized in January 2023, introduces critical updates to Quality Risk Management (QRM) practices, emphasizing the need to address subjectivity, enhance formality, improve risk-based decision-making, and manage product availability risks. These revisions aim to ensure that QRM processes are more science-driven, knowledge-based, and effective in safeguarding product quality and patient safety. Two years later it is important to continue to build on key strategies for reducing subjectivity in QRM and aligning with the updated requirements.

Understanding Subjectivity in QRM

Subjectivity in QRM arises from personal opinions, biases, heuristics, or inconsistent interpretations of risks by stakeholders. This can impact every stage of the QRM process—from hazard identification to risk evaluation and mitigation. The revised ICH Q9(R1) explicitly addresses this issue by introducing a new subsection, “Managing and Minimizing Subjectivity,” which emphasizes that while subjectivity cannot be entirely eliminated, it can be controlled through structured approaches.

The guideline highlights that subjectivity often stems from poorly designed scoring systems, differing perceptions of hazards and risks among stakeholders, and cognitive biases. To mitigate these challenges, organizations must adopt robust strategies that prioritize scientific knowledge and data-driven decision-making.

Strategies to Reduce Subjectivity

Leveraging Knowledge Management

ICH Q9(R1) underscores the importance of knowledge management as a tool to reduce uncertainty and subjectivity in risk assessments. Effective knowledge management involves systematically capturing, organizing, and applying internal and external knowledge to inform QRM activities. This includes maintaining centralized repositories for technical data, fostering real-time information sharing across teams, and learning from past experiences through structured lessons-learned processes.

By integrating knowledge management into QRM, organizations can ensure that decisions are based on comprehensive data rather than subjective estimations. For example, using historical data on process performance or supplier reliability can provide objective insights into potential risks.

To integrate knowledge management (KM) more effectively into quality risk management (QRM), organizations can implement several strategies to ensure decisions are based on comprehensive data rather than subjective estimations:

Establish Robust Knowledge Repositories

Create centralized, easily accessible repositories for storing and organizing historical data, lessons learned, and best practices. These repositories should include:

  • Process performance data
  • Supplier reliability metrics
  • Deviation and CAPA records
  • Audit findings and inspection observations
  • Technology transfer documentation

By maintaining these repositories, organizations can quickly access relevant historical information when conducting risk assessments.

Implement Knowledge Mapping

Conduct knowledge mapping exercises to identify key sources of knowledge within the organization. This process helps to:

Use the resulting knowledge maps to guide risk assessment teams to relevant information and expertise.

Develop Data Analytics Capabilities

Invest in data analytics tools and capabilities to extract meaningful insights from historical data. For example:

  • Use statistical process control to identify trends in manufacturing performance
  • Apply machine learning algorithms to predict potential quality issues based on historical patterns
  • Utilize data visualization tools to present complex risk data in an easily understandable format

These analytics can provide objective, data-driven insights into potential risks and their likelihood of occurrence.

Integrate KM into QRM Processes

Embed KM activities directly into QRM processes to ensure consistent use of available knowledge:

  • Include a knowledge gathering step at the beginning of risk assessments
  • Require risk assessment teams to document the sources of knowledge used in their analysis
  • Implement a formal process for capturing new knowledge generated during risk assessments

This integration helps ensure that all relevant knowledge is considered and that new insights are captured for future use.

Foster a Knowledge-Sharing Culture

Encourage a culture of knowledge sharing and collaboration within the organization:

  • Implement mentoring programs to facilitate the transfer of tacit knowledge
  • Establish communities of practice around key risk areas
  • Recognize and reward employees who contribute valuable knowledge to risk management efforts

By promoting knowledge sharing, organizations can tap into the collective expertise of their workforce to improve risk assessments.

Implementing Structured Risk-Based Decision-Making

The revised guideline introduces a dedicated section on risk-based decision-making, emphasizing the need for structured approaches that consider the complexity, uncertainty, and importance of decisions. Organizations should establish clear criteria for decision-making processes, define acceptable risk tolerance levels, and use evidence-based methods to evaluate options.

Structured decision-making tools can help standardize how risks are assessed and prioritized. Additionally, calibrating expert opinions through formal elicitation techniques can further reduce variability in judgments.

Addressing Cognitive Biases

Cognitive biases—such as overconfidence or anchoring—can distort risk assessments and lead to inconsistent outcomes. To address this, organizations should provide training on recognizing common biases and their impact on decision-making. Encouraging diverse perspectives within risk assessment teams can also help counteract individual biases.

For example, using cross-functional teams ensures that different viewpoints are considered when evaluating risks, leading to more balanced assessments. Regularly reviewing risk assessment outputs for signs of bias or inconsistencies can further enhance objectivity.

Enhancing Formality in QRM

ICH Q9(R1) introduces the concept of a “formality continuum,” which aligns the level of effort and documentation with the complexity and significance of the risk being managed. This approach allows organizations to allocate resources effectively by applying less formal methods to lower-risk issues while reserving rigorous processes for high-risk scenarios.

For instance, routine quality checks may require minimal documentation compared to a comprehensive risk assessment for introducing new manufacturing technologies. By tailoring formality levels appropriately, organizations can ensure consistency while avoiding unnecessary complexity.

Calibrating Expert Opinions

We need to recognize the importance of expert knowledge in QRM activities, but also acknowledges the potential for subjectivity and bias in expert judgments. We need to ensure we:

  • Implement formal processes for expert opinion elicitation
  • Use techniques to calibrate expert judgments, especially when estimating probabilities
  • Provide training on common cognitive biases and their impact on risk assessment
  • Employ diverse teams to counteract individual biases
  • Regularly review risk assessment outputs for signs of bias or inconsistencies

Calibration techniques may include:

  • Structured elicitation protocols that break down complex judgments into more manageable components
  • Feedback and training to help experts align their subjective probability estimates with actual frequencies of events
  • Using multiple experts and aggregating their judgments through methods like Cooke’s classical model
  • Employing facilitation techniques to mitigate groupthink and encourage independent thinking

By calibrating expert opinions, organizations can leverage valuable expertise while minimizing subjectivity in risk assessments.

Utilizing Cooke’s Classical Model

Cooke’s Classical Model is a rigorous method for evaluating and combining expert judgments to quantify uncertainty. Here are the key steps for using the Classical Model to evaluate expert judgment:

Select and calibrate experts:
    • Choose 5-10 experts in the relevant field
    • Have experts assess uncertain quantities (“calibration questions”) for which true values are known or will be known soon
    • These calibration questions should be from the experts’ domain of expertise
    Elicit expert assessments:
      • Have experts provide probabilistic assessments (usually 5%, 50%, and 95% quantiles) for both calibration questions and questions of interest
      • Document experts’ reasoning and rationales
      Score expert performance:
      • Evaluate experts on two measures:
        a) Statistical accuracy: How well their probabilistic assessments match the true values of calibration questions
        b) Informativeness: How precise and focused their uncertainty ranges are
      Calculate performance-based weights:
        • Derive weights for each expert based on their statistical accuracy and informativeness scores
        • Experts performing poorly on calibration questions receive little or no weight
        Combine expert assessments:
        • Use the performance-based weights to aggregate experts’ judgments on the questions of interest
        • This creates a “Decision Maker” combining the experts’ assessments
        Validate the combined assessment:
        • Evaluate the performance of the weighted combination (“Decision Maker”) using the same scoring as for individual experts
        • Compare to equal-weight combination and best-performing individual experts
        Conduct robustness checks:
        • Perform cross-validation by using subsets of calibration questions to form weights
        • Assess how well performance on calibration questions predicts performance on questions of interest

        The Classical Model aims to create an optimal aggregate assessment that outperforms both equal-weight combinations and individual experts. By using objective performance measures from calibration questions, it provides a scientifically defensible method for evaluating and synthesizing expert judgment under uncertainty.

        Using Data to Support Decisions

        ICH Q9(R1) emphasizes the importance of basing risk management decisions on scientific knowledge and data. The guideline encourages organizations to:

        • Develop robust knowledge management systems to capture and maintain product and process knowledge
        • Create standardized repositories for technical data and information
        • Implement systems to collect and convert data into usable knowledge
        • Gather and analyze relevant data to support risk-based decisions
        • Use quantitative methods where feasible, such as statistical models or predictive analytics

        Specific approaches for using data in QRM may include:

        • Analyzing historical data on process performance, deviations, and quality issues to inform risk assessments
        • Employing statistical process control and process capability analysis to evaluate and monitor risks
        • Utilizing data mining and machine learning techniques to identify patterns and potential risks in large datasets
        • Implementing real-time data monitoring systems to enable proactive risk management
        • Conducting formal data quality assessments to ensure decisions are based on reliable information

        Digitalization and emerging technologies can support data-driven decision making, but remember that validation requirements for these technologies should not be overlooked.

        Improving Risk Assessment Tools

        The design of risk assessment tools plays a critical role in minimizing subjectivity. Tools with well-defined scoring criteria and clear guidance on interpreting results can reduce variability in how risks are evaluated. For example, using quantitative methods where feasible—such as statistical models or predictive analytics—can provide more objective insights compared to qualitative scoring systems.

        Organizations should also validate their tools periodically to ensure they remain fit-for-purpose and aligned with current regulatory expectations.

        Leverage Good Risk Questions

        A well-formulated risk question can significantly help reduce subjectivity in quality risk management (QRM) activities. Here’s how a good risk question contributes to reducing subjectivity:

        Clarity and Focus

        A good risk question provides clarity and focus for the risk assessment process. By clearly defining the scope and context of the risk being evaluated, it helps align all participants on what specifically needs to be assessed. This alignment reduces the potential for individual interpretations and subjective assumptions about the risk scenario.

        Specific and Measurable Terms

        Effective risk questions use specific and measurable terms rather than vague or ambiguous language. For example, instead of asking “What are the risks to product quality?”, a better question might be “What are the potential causes of out-of-specification dissolution results for Product X in the next 6 months?”. The specificity in the latter question helps anchor the assessment in objective, measurable criteria.

        Factual Basis

        A well-crafted risk question encourages the use of factual information and data rather than opinions or guesses. It should prompt the risk assessment team to seek out relevant data, historical information, and scientific knowledge to inform their evaluation. This focus on facts and evidence helps minimize the influence of personal biases and subjective judgments.

        Standardized Approach

        Using a consistent format for risk questions across different assessments promotes a standardized approach to risk identification and analysis. This consistency reduces variability in how risks are framed and evaluated, thereby decreasing the potential for subjective interpretations.

        Objective Criteria

        Good risk questions often incorporate or imply objective criteria for risk evaluation. For instance, a question like “What factors could lead to a deviation from the acceptable range of 5-10% for impurity Y?” sets clear, objective parameters for the assessment, reducing the room for subjective interpretation of what constitutes a significant risk.

        Promotes Structured Thinking

        Well-formulated risk questions encourage structured thinking about potential hazards, their causes, and consequences. This structured approach helps assessors focus on objective factors and causal relationships rather than relying on gut feelings or personal opinions.

        Facilitates Knowledge Utilization

        A good risk question should prompt the assessment team to utilize available knowledge effectively. It encourages the team to draw upon relevant data, past experiences, and scientific understanding, thereby grounding the assessment in objective information rather than subjective impressions.

        By crafting risk questions that embody these characteristics, QRM practitioners can significantly reduce the subjectivity in risk assessments, leading to more reliable, consistent, and scientifically sound risk management decisions.

        Fostering a Culture of Continuous Improvement

        Reducing subjectivity in QRM is an ongoing process that requires a commitment to continuous improvement. Organizations should regularly review their QRM practices to identify areas for enhancement and incorporate feedback from stakeholders. Investing in training programs that build competencies in risk assessment methodologies and decision-making frameworks is essential for sustaining progress.

        Moreover, fostering a culture that values transparency, collaboration, and accountability can empower teams to address subjectivity proactively. Encouraging open discussions about uncertainties or disagreements during risk assessments can lead to more robust outcomes.

        Conclusion

        The revisions introduced in ICH Q9(R1) represent a significant step forward in addressing long-standing challenges associated with subjectivity in QRM. By leveraging knowledge management, implementing structured decision-making processes, addressing cognitive biases, enhancing formality levels appropriately, and improving risk assessment tools, organizations can align their practices with the updated guidelines while ensuring more reliable and science-based outcomes.

        It has been two years, it is long past time be be addressing these in your risk management process and quality system.

        Ultimately, reducing subjectivity not only strengthens compliance with regulatory expectations but also enhances the quality of pharmaceutical products and safeguards patient safety—a goal that lies at the heart of effective Quality Risk Management.

        FDA Continues the Discussion on AI/ML

        Many of our organizations are somewhere in the journey of using AI/ML some where in the drug product lifecycle, so it is no surprise that the FDA is continuing the dialogue with the recently published draft of “Considerations for the Use of Artificial Intelligence to Support Regulatory Decision-Making for Drug and Biological Products.”

        This draft guidance lays out a solid approach by using a risk-based credibility assessment framework to establish and evaluate the credibility of AI models. This involves:

        • Determining if the model is adequate for the intended use
        • Defining the question of interest the AI model will address
        • Defining the context of use for the AI model
        • Assessing the AI model risk based on model influence and decision consequence
        • Developing a plan to establish model credibility commensurate with the risk
        • Executing the plan and documenting results

        I think may of us are in the midst of figuring out how to provide sufficient transparency around AI model development, evaluation, and outputs to support regulatory decision-making and what will be found to be acceptable. This sort of guidance is a good way for the agency to further that discussion and I definitely plan on commenting on this one.

        Photo by HARUN BENLu0130 on Pexels.com

        Measuring the Effectiveness of Risk Analysis in Engaging the Risk Management Decision-Making Process

        Effective risk analysis is crucial for informed decision-making and robust risk management. Simply conducting a risk analysis is not enough; its effectiveness in engaging the risk management decision-making process is paramount. This effectiveness is largely driven by the transparency and documentation of the analysis, which supports both stakeholder and third-party reviews. Let’s explore how we can measure this effectiveness and why it matters.

        The Importance of Transparency and Documentation

        Transparency and documentation form the backbone of an effective risk analysis process. They ensure that the methodology, assumptions, and results of the analysis are clear and accessible to all relevant parties. This clarity is essential for:

        1. Building trust among stakeholders
        2. Facilitating informed decision-making
        3. Enabling thorough reviews by internal and external parties
        4. Ensuring compliance with regulatory requirements

        Key Metrics for Measuring Effectiveness

        To gauge the effectiveness of risk analysis in engaging the decision-making process, consider the following metrics:

        1. Stakeholder Engagement Level

        Measure the degree to which stakeholders actively participate in the risk analysis process and utilize its outputs. This can be quantified by:

        • Number of stakeholder meetings or consultations
        • Frequency of stakeholder feedback on risk reports
        • Percentage of stakeholders actively involved in risk discussions

        2. Decision Influence Rate

        Assess how often risk analysis findings directly influence management decisions. Track:

        • Percentage of decisions that reference risk analysis outputs
        • Number of risk mitigation actions implemented based on analysis recommendations

        3. Risk Reporting Quality

        Evaluate the clarity and comprehensiveness of risk reports. Consider:

        • Readability scores of risk documentation
        • Completeness of risk data presented
        • Timeliness of risk reporting

        This is a great place to leverage a rubric.

        4. Third-Party Review Outcomes

        Analyze the results of internal and external audits or reviews:

        • Number of findings or recommendations from reviews
        • Time taken to address review findings
        • Improvement in review scores over time

        5. Risk Analysis Utilization

        Measure how frequently risk analysis tools and outputs are accessed and used:

        • Frequency of access to risk dashboards or reports
        • Number of departments utilizing risk analysis outputs
        • Time spent by decision-makers reviewing risk information

        Implementing Effective Measurement

        To implement these metrics effectively:

        1. Establish Baselines: Determine current performance levels for each metric to track improvements over time.
        2. Set Clear Targets: Define specific, measurable goals for each metric aligned with organizational objectives.
        3. Utilize Technology: Implement risk management software to automate data collection and analysis, improving accuracy and timeliness.
        4. Regular Reporting: Create a schedule for regular reporting of these metrics to relevant stakeholders.
        5. Continuous Improvement: Use the insights gained from these measurements to refine the risk analysis process continually.

        Enhancing Transparency and Documentation

        To improve the effectiveness of risk analysis through better transparency and documentation:

        Standardize Risk Reporting

        Develop standardized templates and formats for risk reports to ensure consistency and completeness. This standardization facilitates easier comparison and analysis across different time periods or business units.

        Implement a Risk Taxonomy

        Create a common language for risk across the organization. A well-defined risk taxonomy ensures that all stakeholders understand and interpret risk information consistently.

        Leverage Visualization Tools

        Utilize data visualization techniques to present risk information in an easily digestible format. Visual representations can make complex risk data more accessible to a broader audience, enhancing engagement in the decision-making process.

        Maintain a Comprehensive Audit Trail

        Document all steps of the risk analysis process, including data sources, methodologies, assumptions, and decision rationales. This audit trail is crucial for both internal reviews and external audits.

        Foster a Culture of Transparency

        Encourage open communication about risks throughout the organization. This cultural shift can lead to more honest and accurate risk reporting, ultimately improving the quality of risk analysis.

        Conclusion

        Measuring the effectiveness of risk analysis in engaging the risk management decision-making process is crucial for organizations seeking to optimize their risk management strategies. By focusing on transparency and documentation, and implementing key metrics to track performance, organizations can ensure that their risk analysis efforts truly drive informed decision-making and robust risk management.

        Remember, the goal is not just to conduct risk analysis, but to make it an integral part of the organization’s decision-making fabric. By continuously measuring and improving the effectiveness of risk analysis, organizations can build resilience, enhance stakeholder trust, and navigate uncertainties with greater confidence.