Fact-checked by Grok 2 weeks ago

Process validation

Process validation is the collection and evaluation of data, from the process design stage through commercial production, which establishes that a process is capable of consistently delivering quality products. This practice is essential in regulated industries such as pharmaceuticals, , and devices to ensure product safety, efficacy, and compliance with standards like those from the U.S. (FDA) and the (ISO). The modern approach to process validation follows a lifecycle model comprising three stages, as outlined in the FDA's 2011 guidance, which updated earlier 1987 principles to incorporate contemporary quality-by-design concepts. Stage 1: involves defining the commercial manufacturing based on knowledge gained from , scale-up activities, and risk analysis to identify critical process parameters and quality attributes. Stage 2: Process Qualification confirms the process's reproducibility, including facility and utility qualification as well as process performance qualification (PPQ) through intensive and testing to characterize variability. Stage 3: Continued Process Verification maintains ongoing assurance of process control during routine commercial production via and periodic reviews. In the context of for medical devices, process validation requires objective evidence that processes—particularly those where output cannot be fully verified by subsequent inspection or —consistently meet predetermined , emphasizing validation protocols, records, and requalification upon changes. As of 2024, the FDA has updated its Quality System Regulation to incorporate :2016, effective February 2, 2026. This framework applies to special processes (e.g., sterilization, ), integrating with broader systems to mitigate risks and ensure . Overall, process validation underscores the principle that quality cannot be adequately assured solely by end-product testing but must be built into the process itself.

Overview

Definition and Principles

Process validation is defined as the collection and evaluation of data, from the process design stage through commercial production, which establishes that a process is capable of consistently delivering quality products that meet predetermined specifications and quality attributes. This documented evidence ensures that manufacturing processes in the pharmaceutical, , and related industries are robust and reliable, minimizing variability and risks to product quality. The core principles of process validation revolve around a lifecycle approach, encompassing process design, qualification, and continued verification to maintain ongoing control. A risk-based methodology is integral, involving the identification and mitigation of potential risks through tools such as quality risk management to prioritize critical aspects of the process. Furthermore, process validation integrates seamlessly with broader quality systems, aligning validation activities with overall quality management practices to support compliance and continuous improvement. Key concepts include the validation master plan (VMP), a high-level document that outlines the scope, responsibilities, protocols, and schedule for validation activities across the facility or . This plan provides a strategic to coordinate efforts and ensure comprehensive coverage. In practice, validation principles emphasize and robustness, with variations depending on the product type; for instance, sterile processes require stringent controls for microbial contamination due to the inability to directly measure sterility in every unit, whereas non-sterile processes, such as oral solid dosage manufacturing, prioritize parameters like uniformity and to ensure batch-to-batch consistency. These examples highlight how validation adapts to inherent process risks while upholding the overarching goal of .

Historical Context and Evolution

Process validation in the emerged in the 1970s as a response to significant failures, particularly terminal sterilization process issues that led to contaminated products and patient harm. This development built on broader regulatory reforms following the crisis of the early , which had prompted the Kefauver-Harris Amendments of 1962 to enhance drug safety and efficacy oversight, indirectly fostering a greater emphasis on . The concept was initially proposed by FDA officials Ted Byers and Bud Loftus to systematically demonstrate that processes reliably produced quality products. The FDA's 1987 Guideline on General Principles of Process Validation marked the first formal regulatory framework, emphasizing prospective validation through installation qualification, operational qualification, and performance qualification to ensure process reproducibility. In the , process validation expanded to address the growing use of computerized systems in drug manufacturing, culminating in the FDA's promulgation of 21 CFR Part 11 in 1997, which established requirements for electronic records and signatures to maintain equivalent to paper systems. This period also saw heightened enforcement, as evidenced by numerous FDA warning letters in 2002 citing validation deficiencies, such as inadequate process controls and documentation failures, which underscored the need for more robust practices across the industry. Concurrently, quality management methodologies like standards, introduced in 1987, and , developed in the mid-1980s, influenced validation by promoting systematic risk-based approaches to variability reduction, extending their application beyond pharmaceuticals to sectors like (via HACCP principles) and medical devices. The evolution toward a more proactive and lifecycle-oriented paradigm accelerated in the 2000s, with the International Council for Harmonisation (ICH) Q8 guideline in 2006 introducing (QbD) principles, which integrated scientific understanding and into development to build quality in from the outset rather than testing it out. This shift was formalized in the FDA's 2011 guidance, which replaced the 1987 document and established a three-stage lifecycle model—, , and continued —moving away from one-time validation events toward ongoing monitoring and adaptation to ensure consistent product quality throughout the .

Regulatory Framework

FDA Lifecycle Approach

The U.S. (FDA) introduced a lifecycle approach to process validation in its 2011 guidance document, "Process Validation: General Principles and Practices," which supersedes the 1987 guideline and shifts emphasis from a traditional batch-based to a - and risk-based model integrated throughout the . This approach applies to the manufacture of human and animal drugs and biological products, promoting the use of knowledge gained during development to ensure consistent product quality, with validation activities spanning design, qualification, and ongoing . The lifecycle consists of three integrated stages. Stage 1, , focuses on developing a process suitable for routine commercial manufacturing through scientific understanding, including identification of critical quality attributes and process parameters via risk assessments and . Stage 2, Process Qualification, confirms the process design's reproducibility under commercial conditions, encompassing facility and equipment qualification as well as process performance qualification through concurrent or prospective runs to demonstrate consistency. Stage 3, Continued Process Verification, provides ongoing assurance that the process remains in control during routine production, involving monitoring, , and adaptation to changes. Regulatory requirements for this approach are grounded in current good manufacturing practices (CGMP) under 21 CFR Parts 210 and 211 for pharmaceuticals, with analogous application to biologics under 21 CFR Parts 600 through 680, ensuring processes consistently meet predetermined specifications. strategies may be used for product families with similar processes to efficiently validate variations while maintaining risk-based justification. In terms of , the lifecycle approach plays a key role in inspections (PAI), where FDA reviewers assess Stage 1 and 2 data to verify process capability before approval, often requiring successful process performance qualification protocols. Inadequate validation can result in serious consequences, such as product recalls, actions, or delays in approval, as failure to ensure process control may lead to quality deviations or adulterated products. This FDA model aligns broadly with international efforts like ICH Q8-Q10 guidelines but maintains a U.S.-centric focus on CGMP .

ICH and International Guidelines

The International Council for Harmonisation (ICH) guidelines Q8, Q9, and Q10 form a foundational framework for process validation, promoting a science- and risk-based approach to pharmaceutical quality. ICH Q9 was revised to Q9(R1) in 2023 to enhance quality risk management throughout the product lifecycle, further supporting process validation strategies. ICH Q8 (Pharmaceutical Development) introduces Quality by Design (QbD), which emphasizes defining critical quality attributes (CQAs) and establishing a design space to ensure robust processes that consistently deliver product quality, thereby supporting flexible validation strategies during scale-up and manufacturing changes. ICH Q9 (Quality Risk Management) integrates risk assessment tools to identify and mitigate risks to CQAs and critical process parameters (CPPs) throughout the validation lifecycle, enabling proactive control and continual improvement. Complementing these, ICH Q10 (Pharmaceutical Quality System) embeds process validation within an overarching quality management system, facilitating knowledge accumulation, change control, and ongoing verification to maintain process performance over time. Together, these guidelines endorse alternative validation approaches, such as continual process verification (CPV), which relies on real-time monitoring rather than solely traditional batch testing, applicable to both drug substances and products. The () aligns closely with ICH principles through its 2014 Guideline on Process Validation for Finished Products (revised in 2016), which adopts a lifecycle approach encompassing , , and continued to demonstrate reproducible quality. This guideline encourages the use of (PAT) and for hybrid or continuous manufacturing, while requiring data from at least three production-scale batches for traditional validation unless justified otherwise. Similarly, the (WHO) reinforces international standards in its Technical Report Series, with guidelines on validation updated in TRS 1019 Annex 3 in 2019, outlining GMP validation principles that include prospective, concurrent, and retrospective methods, with a strong emphasis on risk-based of and processes prior to . WHO guidelines stress documentation, resource allocation, and alignment with ICH for global applicability, particularly in ensuring and lifecycle monitoring. In contrast to the U.S. FDA's three-stage model, ICH and aligned guidelines place greater emphasis on release testing (RTRT), where in-process measurements and enable batch certification without extensive end-product testing, provided risks are managed and validated. Additionally, ICH Q7 ( for Active Pharmaceutical Ingredients) extends validation requirements to , mandating prospective validation of critical steps for new processes—typically involving three consecutive batches—to confirm profiles and before commercial use, with revalidation triggered by significant changes. Global implementation of these guidelines is advanced through mutual recognition agreements (MRAs), such as those between the and partners like the U.S., , and , which enable reliance on foreign GMP inspections and batch certifications, thereby streamlining validation oversight and reducing redundant audits across borders. As of October 1, 2025, the EU-US MRA expanded to include reliance on third-country GMP inspections, enhancing cross-border validation efficiency. However, faces challenges in emerging markets, including divergent interpretations of ICH standards, resource limitations for GMP compliance, and inconsistent trust among regulators, which can delay adoption of lifecycle validation and increase variability in process controls.

Stage 1: Process Design

Critical Quality Attributes (CQA)

Critical Quality Attributes (CQAs) are defined as physical, chemical, biological, or microbiological properties or characteristics of a product that should be within appropriate limits, ranges, or distributions to ensure the desired product quality. These attributes are foundational to pharmaceutical development, as they directly relate to the quality target product profile (QTPP) and must be controlled throughout the manufacturing process to safeguard and product efficacy. The identification of CQAs involves a systematic risk assessment process, often employing tools such as (FMEA) to evaluate potential impacts on product quality. This assessment prioritizes attributes based on their severity of harm to , , and purity if they deviate from specified limits, ensuring that only those with significant risk are designated as critical. For instance, risk evaluation considers how failure in a quality attribute could lead to adverse clinical outcomes, drawing from ICH Q9 principles of quality risk management. Representative examples of CQAs vary by dosage form and therapeutic modality. In oral solid dosage forms like tablets, common CQAs include (active ingredient content), rate, and levels, as these directly influence and . For biologics, such as monoclonal antibodies, key CQAs encompass (biological activity) and aggregation levels, where elevated aggregates pose a high risk of and reduced efficacy. Control strategies for CQAs are established to maintain these attributes within predefined target values and acceptance criteria, forming part of the overall system. These strategies involve monitoring and adjusting process inputs to mitigate variability, with CQAs serving as the primary outputs influenced by critical process parameters (CPPs) to ensure consistent product quality.

Critical Process Parameters (CPP)

Critical Process Parameters (CPPs) are defined as process parameters whose variability has a direct impact on critical quality attributes (CQAs) and whose control is essential for ensuring product quality. This definition, established in the ICH Q8(R2) guideline on pharmaceutical development, emphasizes that CPPs must be monitored or controlled to maintain consistent output that meets predefined quality standards. Unlike non-critical parameters, CPPs are identified based on their potential to influence CQAs, such as purity or potency in drug manufacturing, through and process understanding. Identification of CPPs typically involves multivariate analysis of process data, review of historical manufacturing records, and risk-based evaluations to classify parameters as critical or key. Multivariate techniques, such as , help correlate parameter variability with quality outcomes, while historical data review assesses past performance to pinpoint influential factors. Parameters are classified as critical if their variability significantly affects CQAs, whereas key parameters are important for process efficiency but do not directly impact quality to the same degree. This classification supports targeted control strategies during . Representative examples of CPPs include mixing speed and duration in blending operations, where deviations can lead to content uniformity issues, and and pressure in sterilization processes, which ensure microbial inactivation without compromising product stability. These parameters are selected based on their established links to in specific operations. Control ranges for CPPs are established as proven acceptable ranges (PARs) through risk-based , defining the boundaries within which the parameter can vary—while holding others constant—to consistently achieve acceptable . PARs are derived from and provide a for operational limits, ensuring robustness in .

Design of Experiments (DOE)

() serves as a systematic statistical methodology in the process design stage of validation to investigate the relationships between input variables, such as critical process parameters (CPPs), and output responses, like critical quality attributes (CQAs), thereby defining the design space where quality is assured. This approach enables the identification of how variations in multiple factors simultaneously influence product quality, supporting the establishment of robust process controls. Common types of DOE include full factorial designs, which examine all possible combinations of factor levels to provide comprehensive data on main effects and interactions; fractional factorial designs, which test a subset of combinations to efficiently screen numerous factors when resources are limited; and (RSM), which employs sequential experimentation to model and optimize quadratic relationships between factors and responses for process refinement. These designs are particularly valuable in pharmaceutical development for mapping multivariate interactions that traditional one-factor-at-a-time methods might overlook. Implementation begins with selecting relevant CPPs as factors based on prior risk assessments, defining discrete levels (e.g., low, nominal, high) for each, and determining the number of experimental runs to balance information gain with practicality. Experiments are conducted under controlled conditions, often at lab or pilot scale, followed by statistical analysis using analysis of variance (ANOVA) to quantify main effects, interaction effects, and their significance on responses. For instance, in a two-level , the response Y is typically modeled as Y = \beta_0 + \sum \beta_i X_i + \sum \beta_{ij} X_i X_j + \epsilon where Y represents the measured response, X_i are the coded factor levels, \beta_0 is the intercept, \beta_i and \beta_{ij} are the coefficients for main and interaction effects, respectively, and \epsilon is the random error term; this model facilitates prediction and optimization within the explored space. The primary benefits of DOE lie in its efficiency to minimize trial-and-error experimentation while uncovering critical interactions that ensure process robustness, such as the combined effects of temperature and humidity on drug stability in formulation studies, allowing for targeted control strategies that enhance overall product quality consistency. By providing empirical evidence for parameter ranges, DOE contributes to a science- and risk-based approach to validation, reducing development time and costs in pharmaceutical manufacturing.

Quality by Design (QbD)

Quality by Design (QbD) is a systematic, science-based framework for pharmaceutical development that integrates product and process understanding to ensure quality is built into the manufacturing process from the outset. According to ICH Q8(R2), QbD begins with predefined objectives and relies on prior knowledge, quality risk management, and experimental approaches such as to define a design space where the desired quality is assured. This approach shifts from empirical, trial-and-error methods to a proactive strategy that emphasizes mechanistic understanding of how inputs affect outputs. Key elements of QbD include the Target Product Profile (TPP), which serves as a prospective summary of the quality characteristics—such as , strength, and stability—that a drug product must achieve to meet and goals. Central to this framework are Critical Quality Attributes (CQAs), defined as physical, chemical, biological, or microbiological properties that must remain within specified limits to ensure product quality, such as levels or profiles. Critical Material Attributes (CMAs) refer to properties of excipients, container closure systems, or packaging materials that influence CQAs, like in raw materials. Critical Process Parameters (CPPs) are process variables whose variability impacts CQAs and thus require monitoring or control, such as mixing time or temperature. The knowledge space encompasses the enhanced process understanding derived from systematic studies, while the design space represents the multidimensional range of input variables (CMAs and CPPs) and their interactions that consistently deliver products meeting CQAs; operating within this space does not constitute a change from the approved process. The implementation of QbD follows structured steps to build this understanding. First, the Quality Target Product Profile (QTPP) is defined, outlining the quality criteria needed for the product's intended performance, , and based on clinical and regulatory requirements. Next, CQAs are identified from the QTPP, followed by the determination of CMAs and CPPs through and experimentation to establish links between inputs and quality outcomes. Finally, a control strategy is established, incorporating process controls, specifications, and monitoring to ensure the product remains within the design space and meets quality standards throughout manufacturing. Adopting QbD offers significant advantages, including greater manufacturing flexibility, as adjustments within the established design space do not require regulatory notification or approval. It also reduces the need for post-approval changes by fostering robust product and process knowledge early in development, thereby streamlining regulatory interactions and enhancing overall efficiency.

Process Analytical Technology (PAT)

Process Analytical Technology (PAT) is a framework initiated by the U.S. (FDA) in its 2004 guidance document, designed to enhance the quality of through the integration of timely measurements of critical quality and performance attributes of raw and in-process materials and processes. This approach aims to facilitate innovations in , , and , enabling a shift from traditional end-product testing to a more proactive, science-based system that builds quality directly into the manufacturing process. By emphasizing real-time data acquisition and , PAT supports the identification and of variability to ensure consistent product quality during the process design stage of validation. Key components of PAT include multivariate data acquisition and analysis tools, such as near-infrared () spectroscopy for non-destructive monitoring of chemical and physical attributes, process analyzers that operate at-line, on-line, or in-line to provide measurements, and techniques for extracting meaningful insights from datasets. These elements work together to enable the of robust understanding; for instance, multivariate statistical methods help model multi-factorial relationships in , while process analyzers deliver information on biological, physical, and chemical properties without disrupting operations. , in particular, applies mathematical and statistical to interpret or , supporting predictive modeling for . In the context of process design, PAT applications focus on building empirical models for endpoint detection and optimization, such as determining the optimal granulation endpoint in tablet manufacturing by monitoring moisture content via NIR spectroscopy or assessing sublimation completion in lyophilization through real-time pressure and temperature measurements. These tools integrate seamlessly with (DoE) methodologies, allowing for systematic exploration of critical process parameters to refine models that predict quality outcomes and minimize risks during scale-up. For example, orthogonal experimental designs within PAT frameworks enable the randomization and of variables to establish control strategies early in development. The primary benefits of PAT include a paradigm shift from reliance on post-production testing to continuous, real-time verification, which reduces production cycle times, prevents batch rejects, and lowers reprocessing needs by addressing variability at its source. This continuous quality assurance approach not only improves manufacturing efficiency but also offers regulatory incentives, such as opportunities for reduced end-product batch testing and more flexible approval pathways for well-characterized processes that demonstrate equivalent or superior quality control. Overall, PAT fosters a culture of ongoing knowledge management and process improvement, aligning with broader quality-by-design principles to enhance pharmaceutical validation outcomes.

Stage 2: Process Qualification

Installation Qualification (IQ)

Installation Qualification (IQ) is the documented verification that facilities, utilities, and equipment are designed and installed in accordance with their specifications and the manufacturer's recommendations, ensuring they are suitable for their intended use in the manufacturing process. This phase, part of Stage 2 (Process Qualification) in the FDA's lifecycle approach to process validation, confirms adherence to the User Requirements Specification (URS) and critical aspects such as materials of construction before proceeding to operational testing. Under Good Manufacturing Practice (GMP) guidelines, IQ focuses on objective evidence that the installation is complete and satisfactory, supporting reproducible commercial manufacturing. The IQ protocol typically includes key elements such as certificates for instruments, as-built drawings of the , including manuals and certificates of , and checklists verifying utilities like HVAC systems, , and . It outlines test functions and acceptance criteria based on requirements, including of and software where applicable, component traceability, and material suitability. Purchase specifications, spare parts lists, and configuration details are also documented to ensure all aspects align with design intent. Execution of IQ involves pre-startup inspections to confirm correct per the approved plan, including checks for utilities, services like and wiring, and initial functional tests limited to installation integrity, such as verifying and in cleanrooms or autoclaves. These activities occur after factory acceptance testing () and site acceptance testing (SAT) but before operational qualification, with of measuring devices initiated based on to national or international standards. The IQ process culminates in an IQ report that summarizes results, evaluates any deviations with resolutions, and links back to the , requiring review and approval by the quality unit before releasing systems for further qualification. This documentation provides the foundation for subsequent operational qualification.

Operational Qualification (OQ)

Operational Qualification (OQ) is the documented verification that any new or modified facilities, systems, and equipment operate as intended throughout their anticipated operating ranges, confirming that they are capable of consistently operating within predetermined limits defined by critical process parameters (CPPs). This phase, part of Stage 2: Process Qualification in the FDA lifecycle approach, builds on installation qualification by focusing on functional performance rather than mere setup, ensuring the process can produce results within specified quality attributes under controlled conditions. OQ employs worst-case scenarios to challenge the system, such as testing at maximum and minimum CPPs, to establish confidence in operational reliability before advancing to performance qualification. The scope of OQ encompasses subsystems and components critical to process performance, including utilities like air handling and systems, as well as such as pumps, sensors, and . For similar , bracketing approaches may be applied, where representative units are tested to cover variations, provided scientific justification demonstrates equivalence in . Protocols for OQ involve predefined test plans outlining studies, acceptance criteria, responsibilities, and documentation procedures, often including challenge tests like varying loads in dryers to simulate operational stresses or software validation to confirm functionality under expected inputs and outputs. These tests verify performance during interventions, stoppages, and start-ups, ensuring alignment with process requirements without introducing product-specific variables. Upon completion, OQ generates a summary report compiling test results, data analysis, deviations, and conclusions, which must be reviewed and approved by the quality unit to establish defined operating ranges for subsequent process performance qualification. This report provides objective evidence that the equipment and systems support reproducible manufacturing, serving as a prerequisite for full-scale validation.

Performance Qualification (PQ)

Performance Qualification (PQ), also referred to as Process Performance Qualification (PPQ), is the final phase of process qualification in Stage 2 of , where the commercial-scale manufacturing process is evaluated to confirm its capability for reproducible production of a product meeting all predefined quality requirements. This stage demonstrates that the process performs consistently under normal operating conditions, including variations in raw materials, equipment, environmental factors, and personnel, ensuring the reliability of Critical Quality Attributes (CQAs). PQ builds directly on the results of Operational Qualification (OQ) by extending testing to full commercial batches. The PQ protocol is a detailed, pre-approved that outlines the specific conditions, controls, sampling strategies, testing procedures, and criteria for the validation runs. Protocols can be prospective for newly developed processes, concurrent for ongoing where is collected, or using historical data when justified by prior knowledge and consistency. Sampling plans during PQ are typically more extensive than routine , guided by statistical criteria to ensure representative across the batch, including in-process and end-product testing. A common approach involves at least three consecutive batches at commercial scale to provide sufficient data for statistical analysis and to account for potential variability. Success in PQ is determined by objective criteria, including all CQAs falling within established specifications and evidence of process consistency through statistical metrics. For critical parameters, a (Cpk) of at least 1.33 is widely accepted as indicating a high degree of assurance that the process will produce conforming product, corresponding to fewer than 63 . Deviations from the must be documented, investigated, and justified to confirm they do not impact product quality. Challenges in PQ often arise during scale-up from pilot to full production, where differences in equipment capacity and material flow can affect process performance. For instance, in tablet compression, transitioning to larger-scale rotary presses may lead to variations in tablet weight, hardness, or content uniformity due to changes in flow or die filling dynamics, requiring additional risk assessments and adjustments to maintain CQA consistency.

Stage 3: Continued Process Verification

Monitoring and Control Strategies

Monitoring and control strategies in continued process verification involve the systematic use of statistical tools and (PAT) to detect and manage variability in real-time during routine commercial production, ensuring the process remains in a validated state of control. According to FDA guidance, this stage emphasizes ongoing assurance through data collection and analysis, focusing on critical process parameters (CPPs) and critical quality attributes (CQAs) to maintain product quality and consistency. Key strategies include (SPC) methods such as Shewhart control charts for monitoring individual measurements and detecting large shifts, and (CUSUM) charts for identifying small, sustained process drifts. These tools enable and process capability assessments, supplemented by annual product quality reviews (APQR) that aggregate data from multiple batches to evaluate long-term stability and variability. PAT tools, such as in-line , provide feedback for immediate adjustments, integrating with broader SPC to enhance detection of excursions. Implementation typically begins with defining key performance indicators (KPIs) tied to CPPs and CQAs, such as yield variations or impurity levels, which are tracked via manufacturing execution systems (MES) equipped with automated alerts for deviations beyond control limits. Personnel training in SPC ensures effective data interpretation, with monitoring frequency scaled by risk—daily for high-risk parameters like temperature in sterile processes and monthly for lower-risk attributes. Data from incoming materials, in-process samples, and finished products are routinely analyzed to assess intra- and inter-batch trends, incorporating feedback from production operators to refine controls. In bioprocessing, for instance, of viability using charts monitors performance, alerting to potential depletions that could affect yield, with high-risk parameters checked in real-time to prevent batch failures. This approach not only sustains compliance but also supports continuous improvement by identifying subtle variabilities early.

Change Management and Risk Assessment

Change management in process validation refers to the systematic approach for evaluating, approving, and implementing modifications to validated manufacturing processes to ensure continued product quality and compliance. This involves assessing changes such as equipment upgrades or formulation adjustments using principles from the revised ICH Q9(R1) (2023) Quality Risk Management guideline, which emphasizes science-based risk evaluation tailored to the potential impact on patient safety and product efficacy. The process typically begins with the identification of a proposed change, followed by an impact assessment conducted by a multidisciplinary change control board or expert team to determine the potential effects on critical process parameters (CPPs) and product quality attributes. According to ICH Q10 Pharmaceutical Quality System, changes are evaluated against the established design space and marketing authorization, with quality risk management tools from ICH Q9(R1) integrated to prioritize risks based on severity, occurrence, and detectability. Revalidation triggers are activated for significant changes; for instance, major alterations may necessitate full performance qualification (PQ) or additional process qualification activities, as outlined in the FDA's Process Validation Guidance, which requires re-qualification when changes could affect process stability or capability. Key tools include periodic risk reviews to monitor evolving threats and updates to (FMEA), a method recommended in ICH Q9(R1) for identifying potential failure modes in processes like equipment modifications. Post-change verification protocols, as described in ICH Q10, involve implementation monitoring and confirmation that the change achieves its objectives without unintended quality impacts, often through targeted testing or . In practice, a supplier change for excipients might trigger re-evaluation of CPPs to assess impacts on process consistency, potentially requiring stability studies or batch testing to verify no adverse effects on product . Regulatory reporting is mandatory under 21 CFR 314.70, where major changes to the production demand a prior approval supplement to the FDA before distribution, moderate changes require a 30-day notice supplement, and minor changes are documented in annual reports. These procedures ensure that ongoing monitoring data informs adaptive risk assessments without disrupting steady-state surveillance.

References

  1. [1]
    [PDF] Process Validation: General Principles and Practices | FDA
    Approach to Process Validation. For purposes of this guidance, process validation is defined as the collection and evaluation of data, from the process ...
  2. [2]
    [PDF] Quality System Regulation Process Validation - FDA
    Sep 30, 2015 · Process Validation means establishing by objective evidence that a process consistently produces a result or product meeting its predetermined ...
  3. [3]
    Process validation: Definition & examples ~ What to look out for
    May 11, 2018 · Process validation means establishing by objective evidence that a process consistently produces a result or product meeting its predetermined specifications.
  4. [4]
    ISO 13485:2016 Standard 7.5.6 Validation of processes for ...
    Jun 12, 2018 · Process validation is the systematic planned collection and evaluation of data of processes during the realization, which establishes evidence that a process ...
  5. [5]
    [PDF] A Guide to the Validation Master Plan (VMP) - ISPE
    This article describes the elemental requirements of a Validation. Master Plan. (VMP), what it should look like, what level of detail should be included, and.
  6. [6]
    The History & Future of Validation | Pharmaceutical Engineering - ISPE
    The origins of validation in our industry can be traced to terminal sterilization process failures in the early 1970s.The First 50 Years · Ispe's Contributions · Looking Forward
  7. [7]
    [PDF] A History of the FDA and Drug Regulation in the United States
    Jun 30, 2025 · through FDA's approval process. 1989. The FDA issued guidelines asking drug makers to decide if a drug is likely to have usefulness in ...
  8. [8]
    Overview of Pharmaceutical Process Validation - WebofPharma
    Apr 30, 2025 · The concept of validation was first proposed by two FDA officials, Ted Byers and Bud Loftus, in the mid 1970's in order to improve the quality ...
  9. [9]
    What are FDA Validation Regulations, Standards & Guidelines?
    In May of 1987 the FDA announced its first validation guidance, in the document “Guideline on General Principles of Process Validation (the 1987 guidance)”.History Of Validation · About The Author · Gerry Creaner
  10. [10]
    Computerized Systems in Drug Establishments (2/83) - FDA
    Nov 7, 2014 · This guide discusses some potential problem areas in application of computer systems, provides inspectional guidance, and includes a glossary of terms.
  11. [11]
    Center for Drug Evaluation and Research 2002 - FDA
    Mar 1, 2016 · The Warning Letter cited deficiencies in the firm's Quality and Production systems including a failure to prevent the release of six lots of ...Good Manufacturing Practices... · Inadequate Cleaning... · Fda Finds Cgmp Problems For...
  12. [12]
    [PDF] Q8(R2) - ICH
    In addition, this annex describes the principles of quality by design1 (QbD). The annex is not intended to establish new standards or to introduce new ...
  13. [13]
    Process Validation: General Principles and Practices - FDA
    Aug 24, 2018 · This guidance outlines the general principles and approaches that FDA considers appropriate elements of process validation for the manufacture of human and ...
  14. [14]
    Validation of Cleaning Processes (7/93) - FDA
    Aug 26, 2014 · GUIDE TO INSPECTIONS VALIDATION OF CLEANING PROCESSES. Note: This document is reference material for investigators and other FDA personnel.
  15. [15]
    [PDF] ICH guideline Q8, Q9 and Q10 - questions and answers volume 4
    The objective of process validation are unchanged when using ICH Q8, Q9 and Q10. The main objective of process validation remains that a process.
  16. [16]
    [PDF] Guideline on process validation for finished products
    Nov 21, 2016 · This guideline covers process validation for finished products, including traditional, continuous, hybrid, and design space verification, and ...
  17. [17]
    [PDF] Annex 3 - World Health Organization (WHO)
    Annex 3 provides guidelines on validation, an essential part of good manufacturing practices, covering general principles and qualification. It is not ...
  18. [18]
    Real time release testing - Scientific guideline
    This document outlines the requirements for applications that propose real time release testing for active substances, intermediates and finished products.
  19. [19]
    [PDF] Q7 Good Manufacturing Practice Guidance for Active ... - FDA
    Q7 is Good Manufacturing Practice Guidance for Active Pharmaceutical Ingredients, containing nonbinding recommendations.
  20. [20]
    Mutual recognition agreements (MRA) - European Medicines Agency
    MRAs are trade agreements that aim to facilitate market access and encourage greater international harmonisation of compliance standards while protecting ...
  21. [21]
    Harmonizing health: a global analysis of pharmaceutical regulatory ...
    This global technical harmonization is challenging and takes a long time, so regulatory strengthening activities also encourage convergence of standards and ...
  22. [22]
    [PDF] Q8(R2) Pharmaceutical Development - FDA
    ... 2006 guidance and describes the principles of quality by design (QbD). The Q8(R1) document issued in June 2009 includes the Q8 parent guidance and the annex.
  23. [23]
    Quality by Design I: Application of Failure Mode Effect Analysis ... - NIH
    The FMEA method was used to perform the qualitative risk assessment, which could identify the CQAs that have the greatest chance of causing product failure, ...
  24. [24]
    [PDF] How to Identify Critical Quality Attributes and Critical Process ...
    Oct 6, 2015 · Critical Quality Attributes (CQA) are properties ensuring desired quality. Critical Process Parameters (CPP) are process parameters impacting ...
  25. [25]
    [PDF] ICH guideline Q9 on quality risk management
    Risk assessment. Risk assessment consists of the identification of hazards and the analysis and evaluation of risks associated with exposure to those hazards ...
  26. [26]
    Understanding Pharmaceutical Quality by Design - PMC - NIH
    The quality attributes of a drug product may include identity, assay, content uniformity, degradation products, residual solvents, drug release or dissolution, ...
  27. [27]
    Critical Quality Attributes Assessment and Testing Strategy for ...
    Mar 28, 2019 · Critical Quality Attributes (CQAs) are properties that must be within limits to ensure desired product quality. Their criticality assessment is ...<|control11|><|separator|>
  28. [28]
    Workflow for Criticality Assessment Applied in Biopharmaceutical ...
    Oct 12, 2017 · Identification of critical process parameters that impact product quality is a central task during regulatory requested process validation.
  29. [29]
    [PDF] A Systematic Approach for Selecting Critical Process Parameters for ...
    May 30, 2012 · Critical process parameters (CPPs) and their associated process controls are crucial to drug development, process validation and to the ...
  30. [30]
    Critical process parameters (CPP) — definition and meaning
    Examples of critical process parameters: Temperature: In processes such as drying, granulation and sterilization, the temperature can have a significant effect ...
  31. [31]
    Design of Experiments (DOE): Applications and Benefits in Quality ...
    Fractional factorial designs are less comprehensive than full factorial designs, but they can save time and money. 1.4.3 Response surface methodology (RSM) ...
  32. [32]
    [PDF] FDA Guidance for Industry PAT – A Framework for Innovative ...
    This guidance is intended to describe a regulatory framework (Process Analytical Technology,. PAT) that will encourage the voluntary development and ...
  33. [33]
    [PDF] Process Validation - FDA
    Sep 13, 2023 · ➢Change Management. Page 4. www.fda.gov. Process Validation. PV is defined as the collection and evaluation of data, from the Process Design ...
  34. [34]
    [PDF] General Principles of Software Validation - Final Guidance for ... - FDA
    This document provides guidance on general principles of software validation, including its purpose, scope, and context, for industry and FDA staff.
  35. [35]
    Process Validation for Medical Devices - MasterControl
    Apr 17, 2017 · A process with a "high degree of assurance" of producing conforming product will have a Cpk ≥ 1.33. This means that the nearest specification ...Missing: PQ | Show results with:PQ<|control11|><|separator|>
  36. [36]
    Tableting Scale-Up Challenges - Korsch AG
    Jan 14, 2025 · It is a challenging task to assess the critical quality attributes for large scale productions already during the formulation development stage.
  37. [37]
    Continued Process Verification in Stages 1–3 - ISPE
    A new commercial manufacturing process should go through stage 1, process design; stage 2, process qualification; and stage 3, continued process verification.
  38. [38]
    Continued Process Verification (CPV) – Ongoing Control
    Good CPV favors fit-for-purpose statistics over novelty. Start with Individuals/Moving Range (I-MR) or X̄/R charts depending on subgroup logic; use EWMA or CUSUM ...
  39. [39]
    Continued Process Verification for Bioproduction
    Dec 17, 2020 · Run charts, control charts, and other statistical methods can help monitor biopharmaceutical PQAs as part of continued process verification.Missing: APQR | Show results with:APQR
  40. [40]
    Continued Process Verification Is Critical in Pharmaceutical ...
    Aug 27, 2025 · Regulatory guidelines mandate a three-phase process validation: development, performance qualification, and continued process verification (CPV/ ...
  41. [41]
    [PDF] Quality Risk Management - ICH
    Nov 9, 2005 · This guideline provides principles and examples of tools for quality risk management that can be applied to different aspects of ...
  42. [42]
    None
    ### Summary of Change Management Process from ICH Q10 Guideline
  43. [43]
    21 CFR 314.70 -- Supplements and other changes to an approved ...
    A supplement must be submitted for any change in the drug substance, drug product, production process, quality controls, equipment, or facilities.